11.07.2015 Views

Pathology of the Head and Neck

Pathology of the Head and Neck

Pathology of the Head and Neck

SHOW MORE
SHOW LESS
  • No tags were found...

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Antonio Cardesa · Pieter J. Slootweg (Eds.)<strong>Pathology</strong> <strong>of</strong> <strong>the</strong> <strong>Head</strong> <strong>and</strong> <strong>Neck</strong>


Antonio Cardesa · Pieter J. Slootweg (Eds.)<strong>Pathology</strong><strong>of</strong> <strong>the</strong> <strong>Head</strong> <strong>and</strong> <strong>Neck</strong>With 249 Figures in 308 separate Illustrations<strong>and</strong> 17 Tables123


Pr<strong>of</strong>essor Dr. Antonio CardesaDepartment <strong>of</strong> Pathological AnatomyHospital ClinicUniversity <strong>of</strong> BarcelonaVillarroel 17008036 BarcelonaSpainPr<strong>of</strong>essor Pieter J. SlootwegDepartment <strong>of</strong> <strong>Pathology</strong>University Medical Center St. RadboudP.O. Box 91016500 HB NijmegenThe Ne<strong>the</strong>rl<strong>and</strong>sLibrary <strong>of</strong> Congress Control Number: 2006922731ISBN-10 3-540-30628-5 Springer Berlin Heidelberg New YorkISBN-13 978-3-540-30628-3 Springer Berlin Heidelberg New YorkThis work is subject to copyright. All rights are reserved, whe<strong>the</strong>r <strong>the</strong> whole or part <strong>of</strong> <strong>the</strong> material is concerned,specifically <strong>the</strong> rights <strong>of</strong> translation, reprinting, reuse <strong>of</strong> illustrations, recitation, broadcasting, reproductionon micr<strong>of</strong>ilms or in any o<strong>the</strong>r way <strong>and</strong> storage in data banks. Duplication <strong>of</strong> this publication or parts <strong>the</strong>re<strong>of</strong> ispermitted only under <strong>the</strong> provisions <strong>of</strong> <strong>the</strong> German Copyright Law <strong>of</strong> September 9, 1965, in its current version,<strong>and</strong> permission for use must always be obtained from Springer-Verlag. Violations are liable for prosecutionunder German Copyright Law.Springer is a part <strong>of</strong> Springer Science+Business Mediaspringer.com© Springer-Verlag Berlin Heidelberg 2006Printed in GermanyThe use <strong>of</strong> general descriptive names, trademarks, etc. in this publication does not imply, even in <strong>the</strong> absence <strong>of</strong>a specific statement, that such names are exempt from <strong>the</strong> relevant protective laws <strong>and</strong> regulations <strong>and</strong> <strong>the</strong>re<strong>of</strong>free for general use.Product liability: <strong>the</strong> publishers cannot guarantee <strong>the</strong> accuracy <strong>of</strong> any information about dosage <strong>and</strong> applicationcontained in this book. In every individual case <strong>the</strong> user must check such information by consulting <strong>the</strong>relevant literature.Editor: Gabriele Schröder, HeidelbergDesk Editor: Ellen Blasig, HeidelbergProduction: LE-T E X, Jelonek, Schmidt & Vöckler GbR, LeipzigTypesetting: Satz-Druck-Service, LeimenCover: Frido Steinen-Broo, eStudio Calamar, SpainPrinted on acid-free paper 24/3100/YL 5 4 3 2 1 0


ToGerhard Seifert <strong>and</strong> to Leslie Michaels,great pioneers <strong>of</strong> <strong>Head</strong> <strong>and</strong> <strong>Neck</strong> <strong>Pathology</strong>in Europe <strong>and</strong> founding members<strong>of</strong> <strong>the</strong> Working Groupon <strong>Head</strong> <strong>and</strong> <strong>Neck</strong> <strong>Pathology</strong><strong>of</strong> <strong>the</strong> European Society <strong>of</strong> <strong>Pathology</strong>.


Foreword<strong>Pathology</strong> <strong>of</strong> <strong>the</strong> <strong>Head</strong> <strong>and</strong> <strong>Neck</strong> is an easy soundingtitle for a complex subject matter. This title st<strong>and</strong>s for anaccumulation <strong>of</strong> diverse diseases occurring in differentorgans whose relationship to each o<strong>the</strong>r consists in <strong>the</strong>fact that <strong>the</strong>y are located between <strong>the</strong> base <strong>of</strong> <strong>the</strong> skull<strong>and</strong> <strong>the</strong> thoracic aperture. One reason for assemblingall <strong>the</strong>se different organs under <strong>the</strong> title “<strong>Pathology</strong> <strong>of</strong><strong>the</strong> <strong>Head</strong> <strong>and</strong> <strong>Neck</strong>” is that <strong>the</strong> proximity <strong>of</strong> <strong>the</strong> organs<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck region makes it difficult for <strong>the</strong>surgical pathologist to focus on one <strong>of</strong> <strong>the</strong>se organs <strong>and</strong>neglect <strong>the</strong> pathology <strong>of</strong> o<strong>the</strong>rs, which are only a centimetreapart. A second reason, however, is that <strong>the</strong> upperdigestive tract <strong>and</strong> <strong>the</strong> upper respiratory tract, whichmeet in <strong>the</strong> larynx, have some basic diseases in common,notably squamous cell carcinoma. Thus pathology<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck is both an arbitrary compilation <strong>of</strong>diseases <strong>and</strong>, at least to some extent, a group <strong>of</strong> diseaseentities with a common morphological <strong>and</strong> pathogenetictrunk.The past years have seen remarkable advances inmany fields <strong>of</strong> pathology, including that <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck. There is a need for a book that integrates surgicalpathology with molecular genetics, epidemiology, clinicalbehaviour <strong>and</strong> biology. This book provides a comprehensivedescription <strong>of</strong> <strong>the</strong> manifold aspects <strong>of</strong> <strong>the</strong>morphology <strong>and</strong> pathology <strong>of</strong> <strong>the</strong> organs <strong>of</strong> <strong>the</strong> head<strong>and</strong> neck region. These description, as comprehensive as<strong>the</strong>y may be, also show that <strong>the</strong>re are some areas <strong>of</strong> <strong>the</strong>pathology <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck that remain an unexploredworld. Examples include <strong>the</strong> never-ending problem<strong>of</strong> prognostication <strong>of</strong> tumour diseases, <strong>the</strong> pathogeneticsignificance <strong>of</strong> tumour precursor lesions <strong>and</strong><strong>the</strong> validation <strong>of</strong> appropriate sets <strong>of</strong> tumour markers asmeaningful predictors <strong>of</strong> malignancy.The editors <strong>of</strong> <strong>the</strong> book, Pr<strong>of</strong>essor Antonio Cardesa<strong>and</strong> Pr<strong>of</strong>essor Pieter Slootweg, are leading experts in<strong>the</strong> field <strong>of</strong> <strong>the</strong> pathology <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. As such<strong>the</strong>y are <strong>the</strong> main members <strong>of</strong> <strong>the</strong> Working Group on<strong>Pathology</strong> <strong>of</strong> <strong>the</strong> <strong>Head</strong> <strong>and</strong> <strong>Neck</strong> <strong>of</strong> <strong>the</strong> European Society<strong>of</strong> <strong>Pathology</strong>, one <strong>of</strong> <strong>the</strong> first European working groupsto be founded under <strong>the</strong> auspices <strong>of</strong> <strong>the</strong> European Society<strong>of</strong> <strong>Pathology</strong>. In this multi-author book <strong>the</strong> expertise<strong>of</strong> outst<strong>and</strong>ing experts on <strong>the</strong> pathology <strong>of</strong> <strong>the</strong> head<strong>and</strong> neck in Europe is reflected. The chapters are characterisedby <strong>the</strong> desire to correlate pathology with allnecessary information on clinical features, epidemiology,pathogenesis <strong>and</strong> molecular genetics. The authors <strong>of</strong><strong>the</strong>se chapters have not attempted to be encyclopaedic,but ra<strong>the</strong>r have aimed at providing concise, yet adequateknowledge. They are <strong>the</strong>refore to be warmly commendedfor providing us with an excellent book, which willprove useful to surgical pathologists involved in <strong>the</strong> pathology<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck.Kiel, GermanyMarch 2006Günter Klöppel


Contents IXPrefaceThis book was initially conceived as a unitary group <strong>of</strong>chapters on “<strong>Pathology</strong> <strong>of</strong> <strong>the</strong> <strong>Head</strong> <strong>and</strong> <strong>Neck</strong>”, to bepublished in German within <strong>the</strong> series <strong>of</strong> volumes <strong>of</strong>Remmele’s Textbook <strong>of</strong> <strong>Pathology</strong>. From <strong>the</strong> outset, <strong>the</strong>editorial approach was to concentrate on pathologicalentities that are ei<strong>the</strong>r unique to or quite characteristic <strong>of</strong><strong>the</strong> head <strong>and</strong> neck. At <strong>the</strong> same time, we strove to avoidas much as possible unnecessary details on systemicdiseases that, although involving <strong>the</strong> head <strong>and</strong> neckregion, have <strong>the</strong>ir main focus <strong>of</strong> activity in o<strong>the</strong>r organs.Thus, “<strong>Pathology</strong> <strong>of</strong> <strong>the</strong> <strong>Head</strong> <strong>and</strong> <strong>Neck</strong>” encompasses<strong>the</strong> wide range <strong>of</strong> diseases encountered in <strong>the</strong> complexanatomic region extending proximally from <strong>the</strong> frontalsinuses, orbits, ro<strong>of</strong> <strong>of</strong> <strong>the</strong> sphenoidal sinuses <strong>and</strong> clivusto distally <strong>the</strong> upper borders <strong>of</strong> <strong>the</strong> sternal manubrium,clavicles <strong>and</strong> first ribs. This includes <strong>the</strong> eyes, ears , upperaerodigestive tract, salivary gl<strong>and</strong>s, dental apparatus,thyroid <strong>and</strong> parathyroid gl<strong>and</strong>s, as well as all <strong>the</strong>epi<strong>the</strong>lial, fibrous, fatty, muscular, vascular, lymphoid,cartilaginous, osseous <strong>and</strong> neural tissues or structuresrelated to <strong>the</strong>m.The contents have been divided into ten chapters. Thefirst covers <strong>the</strong> spectrum <strong>of</strong> precursor <strong>and</strong> neoplasticlesions <strong>of</strong> <strong>the</strong> squamous epi<strong>the</strong>lium. It is followed bychapters devoted to <strong>the</strong> nasal cavities <strong>and</strong> paranasalsinuses, oral cavity, maxill<strong>of</strong>acial skeleton <strong>and</strong> teeth,salivary gl<strong>and</strong>s, nasopharynx <strong>and</strong> Waldeyer`s ring,larynx <strong>and</strong> hypopharynx, ear <strong>and</strong> temporal bone,neck <strong>and</strong> neck dissection, as well as eye <strong>and</strong> ocularadnexa. The pathology <strong>of</strong> <strong>the</strong> thyroid <strong>and</strong> parathyroidgl<strong>and</strong>s <strong>and</strong> lymph nodes is covered in greater detailelsewhere.Since <strong>the</strong> authors selected for writing <strong>the</strong> differentchapters are international experts <strong>and</strong> members <strong>of</strong> <strong>the</strong>Working Group on <strong>Head</strong> <strong>and</strong> <strong>Neck</strong> <strong>Pathology</strong> <strong>of</strong> <strong>the</strong>European Society <strong>of</strong> <strong>Pathology</strong>, <strong>the</strong> chief editors <strong>of</strong> <strong>the</strong>series, Pr<strong>of</strong>. Wolfgang Remmele, Pr<strong>of</strong>. Hans Kreipe <strong>and</strong>Pr<strong>of</strong>. Günter Klöppel, accepted that all manuscriptsshould be in English. After <strong>the</strong> original texts had beensubmitted, it became clear to <strong>the</strong> editors <strong>and</strong> publisherthat, in addition to <strong>the</strong>ir translation to fit into Remmele’sTextbook, <strong>the</strong> work warranted publication in Englishas a separate book. Therefore, we want to thank <strong>the</strong> chiefeditors <strong>and</strong> <strong>the</strong> publisher Springer for <strong>the</strong>ir stimulatingsupport <strong>and</strong> trust. We add our special thanks to <strong>the</strong> authorswho produced such an excellent work, as well as tothose secretaries, photographers <strong>and</strong> o<strong>the</strong>rs who helped<strong>the</strong>m.Finally, we should like to express our wish that thisbook on “<strong>Pathology</strong> <strong>of</strong> <strong>the</strong> <strong>Head</strong> <strong>and</strong> <strong>Neck</strong>”, <strong>the</strong> firstever written as a joint project by a Working Group <strong>of</strong> <strong>the</strong>European Society <strong>of</strong> <strong>Pathology</strong>, could serve as an examplefor new books written by o<strong>the</strong>r Working Groups.Barcelona, SpainNijmegen, The Ne<strong>the</strong>rl<strong>and</strong>sMarch 2006Pr<strong>of</strong>. Antonio CardesaPr<strong>of</strong>. Pieter J. Slootweg


Contents1 Benign <strong>and</strong> Potentially MalignantLesions <strong>of</strong> <strong>the</strong> Squamous Epi<strong>the</strong>lium<strong>and</strong> Squamous Cell Carcinoma . . . . 1N. Gale, N. Zidar1.1 Squamous Cell Papilloma<strong>and</strong> Related Lesions . . . . . . . . . . . . . 21.1.1 Squamous Cell Papilloma, Verruca Vulgaris,Condyloma Acuminatum<strong>and</strong> Focal Epi<strong>the</strong>lial Hyperplasia . . . . . 21.1.2 Laryngeal Papillomatosis . . . . . . . . . 31.2 Squamous Intraepi<strong>the</strong>lial Lesions (SILS) 41.2.1 General Considerations . . . . . . . . . . 41.2.2 Terminological Problems . . . . . . . . . 41.2.3 Aetiology . . . . . . . . . . . . . . . . . . . 51.2.3.1 Oral Cavity <strong>and</strong> Oropharyn . . . . . . . . . 51.2.3.2 Larynx . . . . . . . . . . . . . . . . . . . . 51.2.4 Clinical Features<strong>and</strong> Macroscopic Appearances . . . . . . . 61.2.4.1 Oral <strong>and</strong> Oropharyngeal Leukoplakia,Proliferative Verrucous Leukoplakia<strong>and</strong> Erythroplakia . . . . . . . . . . . . . . 61.2.4.2 Laryngeal <strong>and</strong> HypopharyngealLeukoplakia <strong>and</strong> Chronic Laryngitis . . . 71.2.5 Histological Classifications . . . . . . . . . 81.2.5.1 WHO Dysplasia System . . . . . . . . . . 81.2.5.2 The Ljubljana Classification . . . . . . . . 91.2.5.3 Comparison Between<strong>the</strong> Ljubljana Classification<strong>and</strong> WHO 2005 Classification . . . . . . . 111.2.6 Biomarkers Related to Malignant Potential<strong>of</strong> SILs Recognised by Auxiliary<strong>and</strong> Advanced Molecular Methods . . . . . 121.2.7 Treatment <strong>and</strong> Prognosis . . . . . . . . . . 121.2.7.1 Oral Cavity <strong>and</strong> Oropharynx . . . . . . . . 121.2.7.2 Larynx . . . . . . . . . . . . . . . . . . . . . 131.3 Invasive Squamous Cell Carcinoma . . . . 131.3.1 Microinvasive SquamousCell Carcinoma . . . . . . . . . . . . . . . 131.3.2 Conventional SquamousCell Carcinoma . . . . . . . . . . . . . . . 131.3.2.1 Aetiology . . . . . . . . . . . . . . . . . . . 141.3.2.2 Pathologic Features . . . . . . . . . . . . . 141.3.2.3 Grading . . . . . . . . . . . . . . . . . . . . 141.3.2.4 Invasive Front . . . . . . . . . . . . . . . . 151.3.2.5 Stromal Reaction . . . . . . . . . . . . . . 151.3.2.6 Differential Diagnosis . . . . . . . . . . . . 151.3.2.7 Treatment <strong>and</strong> Prognosis . . . . . . . . . . 151.3.3 Spindle Cell Carcinoma . . . . . . . . . . . 161.3.3.1 Aetiology . . . . . . . . . . . . . . . . . . . 161.3.3.2 Pathologic Features . . . . . . . . . . . . . 161.3.3.3 Differential Diagnosis . . . . . . . . . . . . 171.3.3.4 Treatment <strong>and</strong> Prognosis . . . . . . . . . . 171.3.4 Verrucous Carcinoma . . . . . . . . . . . . 171.3.4.1 Aetiology . . . . . . . . . . . . . . . . . . . 171.3.4.2 Pathologic Features . . . . . . . . . . . . . 181.3.4.3 Differential Diagnosis . . . . . . . . . . . . 181.3.4.4 Treatment . . . . . . . . . . . . . . . . . . . 181.3.4.5 Prognosis . . . . . . . . . . . . . . . . . . . 191.3.5 Papillary Squamous Cell Carcinoma . . . . 191.3.5.1 Aetiology . . . . . . . . . . . . . . . . . . . 191.3.5.2 Pathologic Features . . . . . . . . . . . . . 191.3.5.3 Differential Diagnosis . . . . . . . . . . . . 201.3.5.4 Treatment <strong>and</strong> Prognosis . . . . . . . . . . 201.3.6 Basaloid Squamous Cell Carcinoma . . . . 201.3.6.1 Aetiology . . . . . . . . . . . . . . . . . . . 201.3.6.2 Pathologic Features . . . . . . . . . . . . . 201.3.6.3 Differential Diagnosis . . . . . . . . . . . . 211.3.6.4 Treatment <strong>and</strong> Prognosis . . . . . . . . . . 211.3.7 Adenoid Squamous Cell Carcinoma . . . . 221.3.7.1 Pathologic Features . . . . . . . . . . . . . 221.3.7.2 Differential Diagnosis . . . . . . . . . . . . 221.3.7.3 Treatment <strong>and</strong> Prognosis . . . . . . . . . . 221.3.8 Adenosquamous Carcinoma . . . . . . . . 231.3.8.1 Aetiology . . . . . . . . . . . . . . . . . . . 231.3.8.2 Pathologic Features . . . . . . . . . . . . . 231.3.8.3 Differential Diagnosis . . . . . . . . . . . . 231.3.8.4 Treatment <strong>and</strong> Prognosis . . . . . . . . . . 241.3.9 Lymphoepi<strong>the</strong>lial Carcinoma . . . . . . . 241.3.9.1 Aetiology . . . . . . . . . . . . . . . . . . . 241.3.9.2 Pathologic Features . . . . . . . . . . . . . 241.3.9.3 Differential Diagnosis . . . . . . . . . . . . 251.3.9.4 Treatment <strong>and</strong> Prognosis . . . . . . . . . . 251.4 Second Primary Tumours . . . . . . . . . . 251.5 Tumour Spread <strong>and</strong> Metastasising . . . . 251.5.1 Invasion <strong>of</strong> Lymphatic<strong>and</strong> Blood Vessels . . . . . . . . . . . . . . 26


XIIContents1.5.2 Perineural Invasion . . . . . . . . . . . . . 261.5.3 Regional Lymph Node Metastases . . . . . 261.5.3.1 Extracapsular Spreadin Lymph Node Metastases . . . . . . . . . 261.5.3.2 Metastases in <strong>the</strong> S<strong>of</strong>t Tissue<strong>of</strong> <strong>the</strong> <strong>Neck</strong> . . . . . . . . . . . . . . . . . . 271.5.4 Distant Metastasis . . . . . . . . . . . . . . 271.5.5 Micrometastasis . . . . . . . . . . . . . . . 271.6 Molecular <strong>Pathology</strong><strong>of</strong> Squamous Cell Carcinoma . . . . . . . 281.6.1 Detecting Tumour Cells . . . . . . . . . . 281.6.2 Clonal Analysis . . . . . . . . . . . . . . . 281.6.3 Assessment <strong>of</strong> Riskfor Malignant Progression . . . . . . . . . 291.6.4 DNA/RNA Pr<strong>of</strong>ilingin Predicting Metastatic Disease . . . . . . 29References . . . . . . . . . . . . . . . . . . 292 Nasal Cavity<strong>and</strong> Paranasal Sinuses . . . . . . . . . . 39A. Cardesa, L. Alos2.1 Introduction . . . . . . . . . . . . . . . . . . 402.1.1 Embryology . . . . . . . . . . . . . . . . . . 402.1.2 Anatomy . . . . . . . . . . . . . . . . . . . . 402.1.3 Histology . . . . . . . . . . . . . . . . . . . . 402.2. Acute <strong>and</strong> Chronic Rhinosinusitis . . . . . 402.2.1 Viral Infections (Common Cold) . . . . . . 402.2.2 Bacterial Infections . . . . . . . . . . . . . . 402.2.3 Allergic Rhinitis . . . . . . . . . . . . . . . 402.2.4 Atrophic Rhinitis . . . . . . . . . . . . . . . 412.2.5 Hypertrophic Rhinitis . . . . . . . . . . . . 412.2.6 Non-Suppurative Chronic Sinusitis . . . . 412.3 Sinonasal Polyps . . . . . . . . . . . . . . . 412.3.1 Allergic Polyposis . . . . . . . . . . . . . . . 412.3.2 Polyposis in Mucoviscidosis . . . . . . . . . 412.3.3 Polyposis in Immotile Cilia Syndrome<strong>and</strong> in Kartagener’s Syndrome . . . . . . . 412.3.4 Antrochoanal Polyps . . . . . . . . . . . . . 412.4 Sinonasal Hamartomatous<strong>and</strong> Teratoid Lesions . . . . . . . . . . . . . 422.4.1 Hamartomas . . . . . . . . . . . . . . . . . 422.4.2 Teratoid Lesions . . . . . . . . . . . . . . . 422.5 Pseudotumours . . . . . . . . . . . . . . . . 432.5.1 Mucocele . . . . . . . . . . . . . . . . . . . . 432.5.2 Organising Haematoma . . . . . . . . . . . 432.5.3 Amyloidosis . . . . . . . . . . . . . . . . . . 432.5.4 Myospherulosis . . . . . . . . . . . . . . . . 432.5.5 Eosinophilic Angiocentric Fibrosis . . . . 432.5.6 Heterotopic Brain Tissue . . . . . . . . . . 432.6 Fungal Diseases . . . . . . . . . . . . . . . . 442.6.1 Aspergillosis . . . . . . . . . . . . . . . . . 442.6.2 Mucormycosis . . . . . . . . . . . . . . . . . 442.6.3 Rhinosporidiosis . . . . . . . . . . . . . . . 442.7 HIV-Related Infections . . . . . . . . . . . 442.8 Mid-Facial NecrotisingGranulomatous Lesions . . . . . . . . . . . 452.8.1 Wegener’s Granulomatosis . . . . . . . . . 452.8.2 Lepromatous Leprosy . . . . . . . . . . . . 452.8.3 Tuberculosis . . . . . . . . . . . . . . . . . . 452.8.4 Sarcoidosis . . . . . . . . . . . . . . . . . . . 452.8.5 Rhinoscleroma . . . . . . . . . . . . . . . . 452.8.6 Leishmaniasis . . . . . . . . . . . . . . . . . 452.8.7 Cocaine Abuse . . . . . . . . . . . . . . . . 462.8.8 Local Steroid Injections . . . . . . . . . . . 462.9 Benign Epi<strong>the</strong>lial Neoplasms . . . . . . . . 462.9.1 Sinonasal Papillomas . . . . . . . . . . . . . 462.9.1.1 Squamous Cell Papilloma . . . . . . . . . . 462.9.1.2 Exophytic Papilloma . . . . . . . . . . . . . 462.9.1.3 Inverted Papilloma . . . . . . . . . . . . . . 462.9.1.4 Oncocytic Papilloma . . . . . . . . . . . . . 472.9.2 Salivary-Type Adenomas . . . . . . . . . . 482.9.3 Pituitary Adenomas . . . . . . . . . . . . . 482.10 Benign SinonasalS<strong>of</strong>t Tissue Neoplasms . . . . . . . . . . . . 482.10.1 Haemangiomas . . . . . . . . . . . . . . . . 482.10.2 Haemangiopericytoma . . . . . . . . . . . . 482.10.3 Solitary Fibrous Tumour . . . . . . . . . . . 482.10.4 Desmoid Fibromatosis . . . . . . . . . . . . 492.10.5 Fibrous Histiocytoma . . . . . . . . . . . . 492.10.6 Leiomyoma . . . . . . . . . . . . . . . . . . 492.10.7 Schwannoma <strong>and</strong> Neur<strong>of</strong>ibroma . . . . . . 492.10.8 Meningioma . . . . . . . . . . . . . . . . . . 502.10.9 Paraganglioma . . . . . . . . . . . . . . . . 502.10.10 Juvenile Angi<strong>of</strong>ibroma . . . . . . . . . . . . 502.11 Malignant Sinonasal Tumours . . . . . . . 502.11.1 Keratinising SquamousCell Carcinoma . . . . . . . . . . . . . . . . 512.11.2 Cylindrical Cell Carcinoma . . . . . . . . . 522.11.3 SinonasalUndifferentiated Carcinoma . . . . . . . . 532.11.4 Small Cell (Neuroendocrine)Carcinoma . . . . . . . . . . . . . . . . . . . 542.11.5 Primary SinonasalNasopharyngeal-TypeUndifferentiated Carcinoma . . . . . . . . 542.11.6 Malignant Melanoma . . . . . . . . . . . . 552.11.7 Olfactory Neuroblastoma . . . . . . . . . . 572.11.8 Primitive Neuroectodermal Tumour . . . . 582.11.9 High-Grade SinonasalAdenocarcinomas . . . . . . . . . . . . . . 582.11.9.1 Intestinal-TypeAdenocarcinoma . . . . . . . . . . . . . . . 582.11.9.2 Salivary-Type High-GradeAdenocarcinoma . . . . . . . . . . . . . . . 60


ContentsXIII2.11.10 Low-Grade SinonasalAdenocarcinomas . . . . . . . . . . . . . . . 602.11.10.1 Non-Salivary-TypeLow-Grade Adenocarcinomas . . . . . . . 602.11.10.2 Salivary-TypeLow-Grade Adenocarcinomas . . . . . . . 612.11.11 Sinonasal Malignant Lymphomas . . . . . 612.11.12 Extramedullary Plasmacytoma . . . . . . 622.11.13 Fibrosarcoma . . . . . . . . . . . . . . . . . 622.11.14 Malignant Fibrous Histiocytoma . . . . . 632.11.15 Leiomyosarcoma . . . . . . . . . . . . . . . 632.11.16 Rhabdomyosarcoma . . . . . . . . . . . . . 632.11.17 Malignant PeripheralNerve Sheath Tumour . . . . . . . . . . . . 632.11.18 Teratocarcinosarcoma . . . . . . . . . . . . 63References . . . . . . . . . . . . . . . . . . 643 Oral Cavity . . . . . . . . . . . . . . . . . 72J.W. Eveson3.1 Embryonic Rests <strong>and</strong> Heterotopias . . . . 723.1.1 Fordyce Granules/Spots . . . . . . . . . . . 723.1.2 Juxtaoral Organ <strong>of</strong> Chievitz . . . . . . . . 723.2. Vesiculo-Bullous Diseases . . . . . . . . . 723.2.1 Herpes Simplex Infections . . . . . . . . . 723.2.2 Chickenpox <strong>and</strong> Herpes Zoster . . . . . . 733.2.3 H<strong>and</strong>-Foot-<strong>and</strong>-Mouth Disease . . . . . . 733.2.4 Herpangina . . . . . . . . . . . . . . . . . . 743.2.5 Pemphigus Vulgaris . . . . . . . . . . . . . 743.2.6 Pemphigus Vegetans . . . . . . . . . . . . . 743.2.7 Paraneoplastic Pemphigus . . . . . . . . . 753.2.8 Mucous Membrane Pemphigoid . . . . . . 753.2.9 Dermatitis Herpetiformis . . . . . . . . . 763.2.10 Linear IgA Disease . . . . . . . . . . . . . . 763.2.11 Ery<strong>the</strong>ma Multiforme . . . . . . . . . . . . 773.3 Ulcerative Lesions . . . . . . . . . . . . . . 773.3.1 Aphthous Stomatitis(Recurrent Aphthous Ulceration) . . . . . 773.3.2 Behçet Disease . . . . . . . . . . . . . . . . 783.3.3 Reiter Disease . . . . . . . . . . . . . . . . 783.3.4 Median Rhomboid Glossitis . . . . . . . . 783.3.5 Eosinophilic Ulcer(Traumatic Ulcerative Granulomawith Stromal Eosinophilia) . . . . . . . . . 793.3.6 Acute NecrotisingUlcerative Gingivitis . . . . . . . . . . . . . 793.3.7 Wegener’s Granulomatosis . . . . . . . . . 803.3.8 Tuberculosis . . . . . . . . . . . . . . . . . 813.4 White Lesions . . . . . . . . . . . . . . . . 813.4.1 C<strong>and</strong>idosis . . . . . . . . . . . . . . . . . . 813.4.2 Lichen Planus . . . . . . . . . . . . . . . . . 823.4.3 Lupus Ery<strong>the</strong>matosus . . . . . . . . . . . . 833.4.4 Oral Epi<strong>the</strong>lial Naevi . . . . . . . . . . . . 843.4.5 Smoker’s Keratosis . . . . . . . . . . . . . . 843.4.6 Stomatitis Nicotina . . . . . . . . . . . . . 843.4.7 Hairy Tongue . . . . . . . . . . . . . . . . . 853.4.8 Hairy Leukoplakia . . . . . . . . . . . . . . 853.4.9 Geographic Tongue . . . . . . . . . . . . . 853.4.10 Frictional Keratosis . . . . . . . . . . . . . 863.5 Pigmentations . . . . . . . . . . . . . . . . 863.5.1 Amalgam Tattoo . . . . . . . . . . . . . . . 863.5.2 Localised Melanotic Pigmentation . . . . 863.5.2.1 Oral Melanotic Macules . . . . . . . . . . . 863.5.2.2 Melanoacanthoma . . . . . . . . . . . . . . 873.5.2.3 Pigmented Naevi . . . . . . . . . . . . . . . 873.5.3 Premalignant Oral Melanoses<strong>and</strong> Oral Melanoma . . . . . . . . . . . . . 873.5.4 Addison Disease . . . . . . . . . . . . . . . 883.5.5 Peutz Jeghers Syndrome . . . . . . . . . . . 893.5.6 Racial Pigmentation . . . . . . . . . . . . . 893.5.7 Laugier Hunziker Syndrome . . . . . . . . 893.5.8 Smoker’s Melanosis . . . . . . . . . . . . . 893.5.9 Drug-AssociatedOral Pigmentation . . . . . . . . . . . . . 903.6 Hyperplastic Lesions . . . . . . . . . . . . 903.6.1 Fibrous Hyperplasias . . . . . . . . . . . . 903.6.2 Papillary Hyperplasia . . . . . . . . . . . . 903.6.3 Generalised GingivalFibrous Hyperplasia . . . . . . . . . . . . . 913.6.4 Crohn’s Disease . . . . . . . . . . . . . . . 913.6.5 Or<strong>of</strong>acial Granulomatosis . . . . . . . . . 923.6.6 Chronic Marginal Gingivitis<strong>and</strong> Localised GingivalFibrous Hyperplasia . . . . . . . . . . . . . 923.6.7 Peripheral Giant Cell Granuloma(Giant Cell Epulis) . . . . . . . . . . . . . . 933.6.8 Pyogenic Granuloma . . . . . . . . . . . . 933.6.9 Pulse (Vegetable) Granuloma . . . . . . . 933.7 Benign Tumours<strong>and</strong> Pseudotumours . . . . . . . . . . . . . 943.7.1 Giant Cell Fibroma . . . . . . . . . . . . . 943.7.2 Lingual Thyroid . . . . . . . . . . . . . . . 943.7.3 Verruciform Xanthoma . . . . . . . . . . . 953.7.4 Haemangiomas . . . . . . . . . . . . . . . . 953.7.5 Lymphangioma . . . . . . . . . . . . . . . . 953.7.6 Benign Nerve Sheath Tumours . . . . . . . 953.7.6.1 Neur<strong>of</strong>ibroma . . . . . . . . . . . . . . . . 963.7.6.2 Schwannoma . . . . . . . . . . . . . . . . . 963.7.6.3 Neur<strong>of</strong>ibromatosis . . . . . . . . . . . . . . 963.7.6.4 Multiple Neuromasin Endocrine Neoplasia Syndrome . . . . 963.7.7 Granular Cell Tumour(Granular Cell Myoblastoma) . . . . . . . 963.8 Squamous Cell Carcinoma . . . . . . . . . 963.8.1 Introduction . . . . . . . . . . . . . . . . . 963.8.2 Clinical Features . . . . . . . . . . . . . . . 973.8.2.1 Buccal Mucosa . . . . . . . . . . . . . . . . 973.8.2.2 Tongue . . . . . . . . . . . . . . . . . . . . 973.8.2.3 Floor <strong>of</strong> Mouth . . . . . . . . . . . . . . . . 97


XIVContents3.8.2.4 Gingiva <strong>and</strong> Alveolar Ridge . . . . . . . . 973.8.2.5 Hard Palate . . . . . . . . . . . . . . . . . . 983.8.2.6 Retromolar Trigone . . . . . . . . . . . . . 983.8.3 Staging . . . . . . . . . . . . . . . . . . . . 98References . . . . . . . . . . . . . . . . . . 984 Maxill<strong>of</strong>acial Skeleton<strong>and</strong> Teeth . . . . . . . . . . . . . . . . . 104P.J. Slootweg4.1 Introduction . . . . . . . . . . . . . . . . 1044.1.1 Embryology . . . . . . . . . . . . . . . . 1044.1.2 Tooth Development . . . . . . . . . . . . 1044.2 Inflammatory Diseases<strong>of</strong> <strong>the</strong> Maxill<strong>of</strong>acial Bones . . . . . . . . 1044.3 Cysts <strong>of</strong> <strong>the</strong> Jaws . . . . . . . . . . . . . . 1054.3.1 Odontogenic Cysts –Inflammatory w . . . . . . . . . . . . . . 1054.3.1.1 Radicular Cyst . . . . . . . . . . . . . . . 1054.3.1.2 Paradental Cyst . . . . . . . . . . . . . . 1064.3.2 Odontogenic Cysts –Developmental . . . . . . . . . . . . . . . 1064.3.2.1 Dentigerous Cyst . . . . . . . . . . . . . . 1064.3.2.2 Lateral Periodontal Cyst . . . . . . . . . 1074.3.2.3 Gl<strong>and</strong>ular Odontogenic Cyst . . . . . . . 1074.3.2.4 Odontogenic Keratocyst . . . . . . . . . 1074.3.2.5 Gingival Cyst . . . . . . . . . . . . . . . . 1084.3.3 Non-Odontogenic Cysts . . . . . . . . . 1094.3.3.1 Nasopalatine Duct Cyst . . . . . . . . . . 1094.3.3.2 Nasolabial Cyst . . . . . . . . . . . . . . . 1094.3.3.3 Surgical Ciliated Cyst . . . . . . . . . . . 1094.3.4 Pseudocysts . . . . . . . . . . . . . . . . . 1094.3.4.1 Solitary Bone Cyst . . . . . . . . . . . . . 1094.3.4.2 Focal Bone Marrow Defect . . . . . . . . 1094.4 Odontogenic Tumours . . . . . . . . . . 1094.4.1 Odontogenic Tumours –Epi<strong>the</strong>lial . . . . . . . . . . . . . . . . . . 1104.4.1.1 Ameloblastoma . . . . . . . . . . . . . . . 1104.4.1.2 Calcifying Epi<strong>the</strong>lialOdontogenic Tumour . . . . . . . . . . . 1124.4.1.3 Adenomatoid Odontogenic Tumour . . 1124.4.1.4 Squamous Odontogenic Tumour . . . . 1134.4.2 Odontogenic Tumours –Mesenchymal . . . . . . . . . . . . . . . . 1144.4.2.1 Odontogenic Myxoma . . . . . . . . . . . 1144.4.2.2 Odontogenic Fibroma . . . . . . . . . . . 1154.4.2.3 Cementoblastoma . . . . . . . . . . . . . 1164.4.3 Odontogenic Tumours –Mixed Epi<strong>the</strong>lial <strong>and</strong> Mesenchymal . . . 1174.4.3.1 Ameloblastic Fibroma . . . . . . . . . . . 1174.4.3.2 Ameloblastic Fibro-Odontoma . . . . . . 1174.4.3.3 Odontoma – Complex Type . . . . . . . 1184.4.3.4 Odontoma – Compound Type . . . . . . 1184.4.3.5 Odonto-Ameloblastoma . . . . . . . . . 1184.4.3.6 Calcifying Odontogenic Cyst . . . . . . 1184.4.4 Odontogenic Tumours –Malignant . . . . . . . . . . . . . . . . . . 1194.4.4.1 Malignant Ameloblastoma . . . . . . . . 1194.4.4.2 Ameloblastic Carcinoma . . . . . . . . . 1194.4.4.3 Primary Intraosseous Carcinoma . . . . 1194.4.4.4 Clear Cell Odontogenic Carcinoma . . . 1204.4.4.5 Malignant Epi<strong>the</strong>lial OdontogenicGhost Cell Tumour . . . . . . . . . . . . 1204.4.4.6 Odontogenic Sarcoma . . . . . . . . . . . 1204.5 Fibro-Osseous Lesions . . . . . . . . . . 1214.5.1 Fibrous Dysplasia . . . . . . . . . . . . . 1214.5.2 Ossifying Fibroma . . . . . . . . . . . . . 1214.5.3 Osseous Dysplasia . . . . . . . . . . . . . 1234.6 Giant Cell Lesions . . . . . . . . . . . . . 1244.6.1 Central Giant Cell Granuloma . . . . . . 1244.6.2 Cherubism . . . . . . . . . . . . . . . . . 1244.7 Neoplastic Lesions<strong>of</strong> <strong>the</strong> Maxill<strong>of</strong>acial Bones,Non-Odontogenic . . . . . . . . . . . . . 1254.7.1 Osteoma . . . . . . . . . . . . . . . . . . . 1254.7.2 Chordoma . . . . . . . . . . . . . . . . . . 1254.7.3 Melanotic Neuroectodermal Tumour<strong>of</strong> Infancy . . . . . . . . . . . . . . . . . . 126References . . . . . . . . . . . . . . . . . . 1265 Major <strong>and</strong> MinorSalivary Gl<strong>and</strong>s . . . . . . . . . . . . . 132S. Di Palma, R.H.W. Simpson,A. Skalova, I. Leivo5.1 Introduction . . . . . . . . . . . . . . . . . 1325.1.1 Normal Salivary Gl<strong>and</strong>s . . . . . . . . . . 1325.1.2 Developmental Disorders . . . . . . . . . . 1325.2 Obstructive Disorders . . . . . . . . . . . . 1325.2.1 Mucus Escape Reaction . . . . . . . . . . . 1325.2.2 Chronic Sclerosing Sialadenitis<strong>of</strong> <strong>the</strong> Subm<strong>and</strong>ibular Gl<strong>and</strong>(Küttner Tumour) . . . . . . . . . . . . . . 1335.3. Infections . . . . . . . . . . . . . . . . . . . 1335.3.1 Bacteria, Fungi . . . . . . . . . . . . . . . . 1335.3.2 Viruses . . . . . . . . . . . . . . . . . . . . 1335.4 Miscellaneous InflammatoryDisorders . . . . . . . . . . . . . . . . . . . 1335.5 Miscellaneous Non-InflammatoryDisorders . . . . . . . . . . . . . . . . . . . 1335.5.1 Necrotising Sialometaplasia(Salivary Gl<strong>and</strong> Infarction) . . . . . . . . . 1335.5.2 Sialadenosis . . . . . . . . . . . . . . . . . . 1335.5.3 Adenomatoid Hyperplasia<strong>of</strong> Mucous Salivary Gl<strong>and</strong>s . . . . . . . . . 1345.5.4 Irradiation Changes . . . . . . . . . . . . . 134


ContentsXV5.5.5 Tissue ChangesFollowing Fine Needle Aspiration . . . . . 1345.6 Oncocytic Lesions . . . . . . . . . . . . . . 1345.6.1 Focal <strong>and</strong> Diffuse Oncocytosis . . . . . . . 1345.6.2 Ductal Oncocytosis . . . . . . . . . . . . . 1345.6.3 Multifocal NodularOncocytic Hyperplasia . . . . . . . . . . . 1355.7 Cysts . . . . . . . . . . . . . . . . . . . . . . 1355.7.1 Salivary PolycysticDysgenetic Disease . . . . . . . . . . . . . . 1355.7.2 Mucoceles . . . . . . . . . . . . . . . . . . . 1355.7.3 Simple Salivary Duct Cysts . . . . . . . . . 1355.7.4 Lymphoepi<strong>the</strong>lial Cystic Lesions . . . . . . 1355.7.4.1 Benign Lymphoepi<strong>the</strong>lial Cyst . . . . . . . 1355.7.4.2 Cystic Lymphoid Hyperplasia<strong>of</strong> AIDS . . . . . . . . . . . . . . . . . . . . 1365.7.5 Sclerosing Polycystic Sialadenopathy(Sclerosing Polycystic Adenosis) . . . . . . 1365.7.6 O<strong>the</strong>r Cysts . . . . . . . . . . . . . . . . . . 1375.8 Benign Tumours . . . . . . . . . . . . . . . 1375.8.1 Pleomorphic Adenoma . . . . . . . . . . . 1375.8.1.1 Salivary Gl<strong>and</strong> Anlage Tumour(“Congenital Pleomorphic Adenoma”) . . 1405.8.2 Benign Myoepi<strong>the</strong>lioma . . . . . . . . . . . 1405.8.3 Basal Cell Adenoma . . . . . . . . . . . . . 1415.8.4 Warthin’s Tumour . . . . . . . . . . . . . . 1425.8.5 Oncocytoma . . . . . . . . . . . . . . . . . 1435.8.6 Canalicular Adenoma . . . . . . . . . . . . 1435.8.7 Sebaceous Adenoma . . . . . . . . . . . . . 1445.8.8 Sebaceous Lymphadenoma . . . . . . . . . 1445.8.9 Ductal Papilloma . . . . . . . . . . . . . . . 1445.8.10 Cystadenoma . . . . . . . . . . . . . . . . . 1445.9 Malignant Epi<strong>the</strong>lial Tumours . . . . . . . 1445.9.1 Acinic Cell Carcinoma . . . . . . . . . . . 1445.9.2 Mucoepidermoid Carcinoma . . . . . . . . 1465.9.3 Adenoid Cystic Carcinoma . . . . . . . . . 1475.9.4 Polymorphous Low-GradeAdenocarcinoma . . . . . . . . . . . . . . . 1485.9.4.1 Cribriform Adenocarcinoma<strong>of</strong> <strong>the</strong> Tongue . . . . . . . . . . . . . . . . . 1495.9.5 Epi<strong>the</strong>lial-Myoepi<strong>the</strong>lial Carcinoma . . . 1505.9.6 Hyalinising Clear Cell Carcinoma . . . . 1515.9.7 Basal Cell Adenocarcinoma . . . . . . . . 1515.9.8 Myoepi<strong>the</strong>lial Carcinoma(Malignant Myoepi<strong>the</strong>lioma) . . . . . . . . 1525.9.9 Salivary Duct Carcinoma . . . . . . . . . . 1545.9.10 Oncocytic Carcinoma . . . . . . . . . . . . 1555.9.11 Malignancy in Pleomorphic AdenomaMalignant Mixed Tumour . . . . . . . . . 1565.9.11.1 Carcinoma(True Malignant Mixed Tumour)Ex Pleomorphic Adenoma . . . . . . . . . 1565.9.11.2 CarcinosarcomaEx Pleomorphic Adenoma . . . . . . . . . 1575.9.11.3 Metastasising Pleomorphic Adenoma . . . 1575.9.12 Sebaceous Carcinoma . . . . . . . . . . . . 1585.9.13 Lymphoepi<strong>the</strong>lial Carcinoma . . . . . . . 1585.9.14 Small Cell Carcinoma . . . . . . . . . . . . 1585.9.15 Higher Grade Change in Carcinomas . . . 1595.9.16 Metastatic Malignancies . . . . . . . . . . 1595.10 Hybrid Carcinoma . . . . . . . . . . . . . . 1605.11 Endodermal Sinus Tumour . . . . . . . . . 1605.12 Sialoblastoma . . . . . . . . . . . . . . . . . 1605.13 Alterations in Gene Expression<strong>and</strong> Molecular Derangementsin Salivary Gl<strong>and</strong> Carcinoma . . . . . . . . 1605.13.1 PredominantlyMyoepi<strong>the</strong>lial Malignancies . . . . . . . . . 1615.13.2 PredominantlyEpi<strong>the</strong>lial Malignancies . . . . . . . . . . . 1615.14 Benign <strong>and</strong> MalignantLymphoid Infiltrates . . . . . . . . . . . . . 1625.14.1 Non-AutoimmuneLymphoid Infiltrates . . . . . . . . . . . . . 1625.14.2 Benign AutoimmuneLymphoid Infiltrates . . . . . . . . . . . . . 1625.14.3 Malignant Lymphoma . . . . . . . . . . . . 1635.15 O<strong>the</strong>r Tumours . . . . . . . . . . . . . . . . 1635.16 Unclassified Tumours . . . . . . . . . . . . 163References . . . . . . . . . . . . . . . . . . . 1646 Nasopharynx<strong>and</strong> Waldeyer’s Ring . . . . . . . . . . . 171S. Regauer6.1 Embryological Development<strong>of</strong> <strong>the</strong> Nasopharynx<strong>and</strong> Waldeyer’s Ring . . . . . . . . . . . . . 1726.2 Nasopharynx . . . . . . . . . . . . . . . . . 1736.2.1 Anatomy <strong>and</strong> Histology . . . . . . . . . . . 1736.2.2 Congenital DevelopmentalAnomalies . . . . . . . . . . . . . . . . . . 1736.2.2.1 NasopharyngealBranchial Cleft Cysts . . . . . . . . . . . . 1736.2.2.2 Tornwaldt’s Cyst . . . . . . . . . . . . . . . 1736.2.2.3 Rathke’s Cleft Cyst/Ectopic Pituitary Tissue . . . . . . . . . . . 1746.2.2.4 Craniopharyngioma . . . . . . . . . . . . . 1746.2.2.5 Heterotopic Brain Tissue/Encephalocele . . . . . . . . . . . . . . . . 1746.2.3 Congenital Tumours . . . . . . . . . . . . . 1746.2.3.1 Salivary Gl<strong>and</strong> Anlage Tumour . . . . . . 1756.2.3.2 Hairy Polyp . . . . . . . . . . . . . . . . . . 1756.2.3.3 Congenital NasopharyngealTeratoma . . . . . . . . . . . . . . . . . . . 1756.2.4 Benign Tumours<strong>and</strong> Tumour-Like Lesions . . . . . . . . . 175


XVIContents6.2.4.1 Nasopharyngeal Angi<strong>of</strong>ibroma . . . . . . 1756.2.4.2 Respiratory Epi<strong>the</strong>lialAdenomatoid Hamartoma . . . . . . . . . 1786.2.4.3 Nasopharyngeal Inverted Papilloma . . . 1786.2.4.4 Solitary Fibrous Tumour . . . . . . . . . . 1796.2.4.5 Paraganglioma . . . . . . . . . . . . . . . . 1796.2.4.6 Meningioma . . . . . . . . . . . . . . . . . 1796.2.4.7 Gl<strong>and</strong>ular Retention Cysts . . . . . . . . . 1796.2.5 Nasopharyngeal Carcinoma . . . . . . . . 1806.2.5.1 Non-Keratinising NasopharyngealCarcinoma . . . . . . . . . . . . . . . . . . 1806.2.5.2 Keratinising NasopharyngealCarcinoma . . . . . . . . . . . . . . . . . . . 1826.2.6 Nasopharyngeal Adenocarcinoma . . . . . 1826.2.6.1 Salivary Gl<strong>and</strong>-Type Adenocarcinoma<strong>of</strong> <strong>the</strong> Nasopharynx . . . . . . . . . . . . . 1826.2.6.2 Papillary Adenocarcinoma<strong>of</strong> <strong>the</strong> Nasopharynx . . . . . . . . . . . . . 1826.2.7 Malignant Non-Epi<strong>the</strong>lial Tumours<strong>of</strong> <strong>the</strong> Nasopharynx . . . . . . . . . . . . . 1836.2.7.1 Chordoma . . . . . . . . . . . . . . . . . . . 1836.2.7.2 Sarcoma . . . . . . . . . . . . . . . . . . . . 1836.3 Waldeyer’s Ring . . . . . . . . . . . . . . . 1836.3.1 Anatomy <strong>and</strong> Histology<strong>of</strong> Waldeyer’s Ring . . . . . . . . . . . . . . 1836.3.2 Congenital Anomalies<strong>of</strong> Waldeyer’s Ring . . . . . . . . . . . . . . 1846.3.3 Tonsillitis . . . . . . . . . . . . . . . . . . . 1846.3.3.1 Bacterial Tonsillitis . . . . . . . . . . . . . 1846.3.3.2 Viral Tonsillitis . . . . . . . . . . . . . . . 1856.3.4 Benign Tumours<strong>of</strong> Waldeyer’s Ring . . . . . . . . . . . . . . 1876.3.4.1 Squamous Papilloma . . . . . . . . . . . . 1876.3.4.2 Lymphangiomatous Tonsillar Polyp . . . . 1876.3.5 Carcinomas<strong>of</strong> Waldeyer’s Ring . . . . . . . . . . . . . . 1876.3.6 Malignant Lymphomas<strong>of</strong> Waldeyer’s Ring . . . . . . . . . . . . . . 1896.3.6.1 Mantle Cell Lymphoma . . . . . . . . . . . 1896.3.6.2 Extranodal Marginal Zone B-CellLymphoma <strong>of</strong> Mucosa-AssociatedLymphoid Tissue . . . . . . . . . . . . . . . 1906.3.6.3 Extranodal NK/T-Cell Lymphoma,Nasal Type . . . . . . . . . . . . . . . . . . 1906.3.6.4 Hodgkin’s Lymphoma . . . . . . . . . . . . 1906.3.6.5 Extramedullary Plasmacytoma . . . . . . 1906.3.7 Systemic DiseaseAffecting Waldeyer’s Ring . . . . . . . . . 190References . . . . . . . . . . . . . . . . . . 1917 Larynx <strong>and</strong> Hypopharynx . . . . . . . 196N. Gale, A. Cardesa, N. Zidar7.1 Summary <strong>of</strong> Anatomy,Histology <strong>and</strong> Embryology . . . . . . . . . 1987.2 Laryngocele, Cysts, Heterotopia . . . . . . 1997.2.1 General Considerations . . . . . . . . . . . 1997.2.2 Laryngocele . . . . . . . . . . . . . . . . . . 1997.2.3 Sacccular Cyst . . . . . . . . . . . . . . . . 1997.2.4 Ductal Cyst . . . . . . . . . . . . . . . . . . 1997.2.5 Oncocytic Cyst . . . . . . . . . . . . . . . . 2007.2.6 Zenker’s Hypopharyngeal Diverticle . . . 2017.2.7 Aberrant Thyroid Tissue . . . . . . . . . . 2017.2.8 Tracheopathia Osteochondroplastica . . . 2027.3 Inflammatory Lesions . . . . . . . . . . . . 2027.3.1 Acute Infections . . . . . . . . . . . . . . . 2027.3.1.1 Epiglottitis . . . . . . . . . . . . . . . . . . 2027.3.1.2 Laryngotracheobronchitis . . . . . . . . . 2027.3.1.3 Diph<strong>the</strong>ria . . . . . . . . . . . . . . . . . . 2027.3.2 Chronic Infections . . . . . . . . . . . . . . 2027.3.2.1 Tuberculosis . . . . . . . . . . . . . . . . . 2027.3.2.2 Fungal Infections . . . . . . . . . . . . . . 2037.3.2.3 O<strong>the</strong>r Rare Infections . . . . . . . . . . . . 2037.3.3 Non-Infectious InflammatoryLesions . . . . . . . . . . . . . . . . . . . . . 2037.3.3.1 Wegener’s Granulomatosis . . . . . . . . . 2037.3.3.2 Sarcoidosis . . . . . . . . . . . . . . . . . . 2047.3.3.3 Rheumatoid Arthritis . . . . . . . . . . . . 2047.3.3.4 Relapsing Polychondritis . . . . . . . . . . 2057.3.3.5 Gout . . . . . . . . . . . . . . . . . . . . . . 2067.3.3.6 Teflon Granuloma . . . . . . . . . . . . . . 2067.3.3.7 Idiopathic SubglotticLaryngeal Stenosis . . . . . . . . . . . . . . 2067.3.3.8 Angioneurotic Oedema . . . . . . . . . . . 2077.4 Degenerative Lesions . . . . . . . . . . . . 2077.4.1 OculopharyngealMuscular Dystrophy . . . . . . . . . . . . . 2077.5 Pseudotumours . . . . . . . . . . . . . . . . 2077.5.1 Exudative Lesions <strong>of</strong> Reinke’s Space . . . . 2077.5.1.1 Reinke’s Oedema . . . . . . . . . . . . . . . 2087.5.1.2 Vocal Cord Polyp <strong>and</strong> Nodule . . . . . . . 2087.5.2 Contact Ulcer <strong>and</strong> Granuloma,Intubation Granuloma . . . . . . . . . . . 2107.5.3 Necrotising Sialometaplasia . . . . . . . . 2117.5.4 Metaplastic Elastic CartilaginousNodules . . . . . . . . . . . . . . . . . . . . 2117.5.5 Amyloidosis . . . . . . . . . . . . . . . . . . 2117.5.6 Sinus Histiocytosiswith Massive Lymphadenopathy<strong>and</strong> O<strong>the</strong>r Rare Pseudotumours . . . . . . 2127.5.7 Inflammatory My<strong>of</strong>ibroblasticTumour . . . . . . . . . . . . . . . . . . . . 2137.6 Benign Neoplasms . . . . . . . . . . . . . . 2147.6.1 Squamous Cell Papilloma . . . . . . . . . . 2147.6.2 Salivary Gl<strong>and</strong>-Type Tumours . . . . . . . 2147.6.2.1 Pleomorphic Adenoma . . . . . . . . . . . 2147.6.2.2 Oncocytoma . . . . . . . . . . . . . . . . . 2147.6.3 Haemangioma(Neonatal <strong>and</strong> Adult Types) . . . . . . . . 214


ContentsXVII7.6.4 Paraganglioma . . . . . . . . . . . . . . . . 2157.6.5 Granular Cell Tumour . . . . . . . . . . . 2167.6.6 Chondroma . . . . . . . . . . . . . . . . . . 2177.7 Malignant Neoplasms . . . . . . . . . . . . 2177.7.1 Potentially Malignant (Precancerous)Lesions . . . . . . . . . . . . . . . . . . . . 2177.7.2 Invasive Squamous Cell Carcinoma . . . . 2187.7.2.1 Epidemiology . . . . . . . . . . . . . . . . . 2187.7.2.2 Aetiology . . . . . . . . . . . . . . . . . . . 2187.7.2.3 Anatomic Sites . . . . . . . . . . . . . . . . 2187.7.2.4 Histological Variants . . . . . . . . . . . . 2197.7.2.5 TNM Grading . . . . . . . . . . . . . . . . 2207.7.3 Neuroendocrine Carcinoma . . . . . . . . 2207.7.3.1 Well-DifferentiatedNeuroendocrine Carcinoma(Carcinoid) . . . . . . . . . . . . . . . . . . 2207.7.3.2 Moderately DifferentiatedNeuroendocrine Carcinoma(Atypical Carcinoid) . . . . . . . . . . . . . 2207.7.3.3 Poorly DifferentiatedNeuroendocrine Carcinoma(Small Cell Carcinoma) . . . . . . . . . . . 2217.7.4 Adenocarcinoma . . . . . . . . . . . . . . . 2227.7.4.1 Adenoid Cystic Carcinoma . . . . . . . . . 2227.7.4.2 Mucoepidermoid Carcinoma . . . . . . . . 2227.7.5 Sarcomas . . . . . . . . . . . . . . . . . . . 2237.7.5.1 Chondrosarcoma . . . . . . . . . . . . . . . 2237.7.5.2 O<strong>the</strong>r Sarcomas . . . . . . . . . . . . . . . 2247.7.6 O<strong>the</strong>r Malignant Neoplasms . . . . . . . . 2247.7.6.1 Malignant Lymphoma . . . . . . . . . . . . 2247.7.6.2 Extraosseus (Extramedullary)Plasmacytoma . . . . . . . . . . . . . . . . 2247.7.6.3 Primary Mucosal Melanoma . . . . . . . . 2257.7.6.4 Metastases to <strong>the</strong> Larynx . . . . . . . . . . 225References . . . . . . . . . . . . . . . . . . . 2268 Ear <strong>and</strong> Temporal Bone . . . . . . . . . 234L. Michaels8.1 Summary <strong>of</strong> Embryology,Anatomy <strong>and</strong> Histology . . . . . . . . . . . 2368.1.1 Embryology . . . . . . . . . . . . . . . . . . 2368.1.2 Anatomy . . . . . . . . . . . . . . . . . . . . 2368.1.3 Histology . . . . . . . . . . . . . . . . . . . 2378.2 External Ear <strong>and</strong> Auditory Canal . . . . . 2378.2.1 Inflammatory <strong>and</strong> Metabolic Lesions . . . 2378.2.1.1 Diffuse External Otitis . . . . . . . . . . . . 2378.2.1.2 Perichondritis . . . . . . . . . . . . . . . . 2378.2.1.3 Malignant Otitis Externa . . . . . . . . . . 2378.2.1.4 Relapsing Polychondritis . . . . . . . . . . 2388.2.1.5 Gout . . . . . . . . . . . . . . . . . . . . . . 2388.2.1.6 Ochronosis . . . . . . . . . . . . . . . . . . 2388.2.2 Pseudocystic <strong>and</strong> Cystic Lesions . . . . . . 2388.2.2.1 Idiopathic PseudocysticChondromalacia . . . . . . . . . . . . . . . 2388.2.2.2 First Branchial Cleft Cyst . . . . . . . . . . 2388.2.3 Tumour-Like Lesions . . . . . . . . . . . . 2398.2.3.1 Chondrodermatitis Nodularis Helicis . . . 2398.2.3.2 Keratosis Obturans <strong>and</strong> Cholesteatoma<strong>of</strong> External Canal . . . . . . . . . . . . . . 2398.2.3.3 Keratin Granuloma . . . . . . . . . . . . . 2398.2.3.4 Angiolymphoid Hyperplasiawith Eosinophilia<strong>and</strong> Kimura’s Disease . . . . . . . . . . . . 2398.2.3.5 Accessory Tragus . . . . . . . . . . . . . . 2408.2.3.6 Keloid . . . . . . . . . . . . . . . . . . . . . 2408.2.4 Benign Neoplasms . . . . . . . . . . . . . . 2408.2.4.1 Adenoma <strong>of</strong> Ceruminal Gl<strong>and</strong>s . . . . . . 2408.2.4.2 Pleomorphic Adenoma<strong>of</strong> Ceruminal Gl<strong>and</strong>s . . . . . . . . . . . . 2418.2.4.3 Syringocystadenoma Papilliferum<strong>of</strong> Ceruminal Gl<strong>and</strong>s . . . . . . . . . . . . 2418.2.4.4 Bony Lesions . . . . . . . . . . . . . . . . . 2418.2.5 Malignant Neoplasms . . . . . . . . . . . . 2428.2.5.1 Adenocarcinoma<strong>of</strong> Ceruminal Gl<strong>and</strong>s . . . . . . . . . . . . 2428.2.5.2 Adenoid Cystic Carcinoma<strong>of</strong> Ceruminal Gl<strong>and</strong>s . . . . . . . . . . . . 2428.2.5.3 Basal Cell Carcinoma . . . . . . . . . . . . 2428.2.5.4 Squamous Cell Carcinoma . . . . . . . . . 2438.2.5.5 Melanotic Neoplasms . . . . . . . . . . . . 2438.3 Middle Ear <strong>and</strong> Mastoid . . . . . . . . . . 2448.3.1 Inflammatory Lesions . . . . . . . . . . . . 2448.3.1.1 Acute <strong>and</strong> Chronic Otitis Media . . . . . . 2448.3.1.2 Cholesteatoma . . . . . . . . . . . . . . . . 2448.3.1.3 Unusual Inflammatory Lesions . . . . . . 2478.3.2 Neoplasms <strong>and</strong> LesionsResembling Neoplasms . . . . . . . . . . . 2478.3.2.1 Choristoma(Salivary Gl<strong>and</strong>, Glial<strong>and</strong> Sebaceous Types) . . . . . . . . . . . . 2478.3.2.2 Adenoma . . . . . . . . . . . . . . . . . . . 2478.3.2.3 Papillary Tumours . . . . . . . . . . . . . . 2488.3.2.4 Jugulotympanic Paraganglioma . . . . . . 2498.3.2.5 Squamous Carcinoma . . . . . . . . . . . . 2508.3.2.6 Meningioma . . . . . . . . . . . . . . . . . 2518.3.2.7 Rhabdomyosarcoma . . . . . . . . . . . . . 2518.3.2.8 Metastatic Carcinoma . . . . . . . . . . . . 2528.4 Inner Ear . . . . . . . . . . . . . . . . . . . 2528.4.1 Bony Labyrinth . . . . . . . . . . . . . . . . 2528.4.1.1 Otosclerosis . . . . . . . . . . . . . . . . . . 2528.4.1.2 Paget’s Disease . . . . . . . . . . . . . . . . 2538.4.1.3 Osteogenesis Imperfecta . . . . . . . . . . 2548.4.1.4 Osteopetrosis . . . . . . . . . . . . . . . . . 2548.4.2 Membranous Labyrinth<strong>and</strong> Cranial Nerves . . . . . . . . . . . . . 2548.4.2.1 Viral, Bacterial<strong>and</strong> Mycotic Infections . . . . . . . . . . . 2548.4.2.2 Lesions <strong>of</strong> <strong>the</strong> Vestibular System . . . . . . 2568.4.2.3 Tumours <strong>and</strong> Tumour-Like Lesions . . . . 257


XVIIIContents8.4.2.4 Presbyacusis . . . . . . . . . . . . . . . . . 2608.4.2.5 Malformations . . . . . . . . . . . . . . . . 260References . . . . . . . . . . . . . . . . . . . 2609 Cysts <strong>and</strong> Unknown Primary<strong>and</strong> Secondary Tumours <strong>of</strong> <strong>the</strong> <strong>Neck</strong>,<strong>and</strong> <strong>Neck</strong> Dissection . . . . . . . . . . . 262M. A Luna, K. Pineda-Daboin9.1 Introduction . . . . . . . . . . . . . . . . . 2649.2. Anatomy . . . . . . . . . . . . . . . . . . . 2649.2.1 Triangles <strong>of</strong> <strong>the</strong> <strong>Neck</strong> . . . . . . . . . . . . 2649.2.2 Lymph Node Regions<strong>of</strong> <strong>the</strong> <strong>Neck</strong> . . . . . . . . . . . . . . . . . . 2649.3 Cysts <strong>of</strong> <strong>the</strong> <strong>Neck</strong> . . . . . . . . . . . . . . 2649.3.1 Developmental Cysts . . . . . . . . . . . . 2659.3.1.1 Branchial Cleft Cysts,Sinuses <strong>and</strong> Fistulae . . . . . . . . . . . . . 2659.3.2 Branchiogenic Carcinoma . . . . . . . . . 2679.3.3 Thyroglossal Duct Cyst<strong>and</strong> Ectopic Thyroid . . . . . . . . . . . . . 2689.3.4 Cervical Thymic Cyst . . . . . . . . . . . . 2699.3.5 Cervical Parathyroid Cyst . . . . . . . . . 2709.3.6 Cervical Bronchogenic Cyst . . . . . . . . 2709.3.7 Dermoid Cyst . . . . . . . . . . . . . . . . 2719.3.8 Unclassified Cervical Cyst . . . . . . . . . 2719.3.9 Non-Developmental Cysts . . . . . . . . . 2719.3.9.1 Ranula . . . . . . . . . . . . . . . . . . . . . 2719.3.9.2 Laryngocele . . . . . . . . . . . . . . . . . . 2719.4 Cystic Neoplasms . . . . . . . . . . . . . . 2729.4.1 Cystic Hygroma<strong>and</strong> Lymphangioma . . . . . . . . . . . . . 2729.4.2 Haemangioma . . . . . . . . . . . . . . . . 2729.4.3 Teratoma . . . . . . . . . . . . . . . . . . . 2729.4.4 Cervical SalivaryGl<strong>and</strong> Cystic Neoplasms . . . . . . . . . . 2739.4.5 Miscellaneous Lesions . . . . . . . . . . . 2739.5 Paraganglioma . . . . . . . . . . . . . . . . 2739.6 Unknown Primary<strong>and</strong> Secondary Tumours . . . . . . . . . . 2749.6.1 Definition . . . . . . . . . . . . . . . . . . . 2749.6.2 Clinical Features . . . . . . . . . . . . . . . 2759.6.3 Search for <strong>the</strong> Primary Tumour . . . . . . 2759.6.4 Common Location<strong>of</strong> <strong>the</strong> Primary Tumour . . . . . . . . . . . 2769.6.5 Histologic Type <strong>of</strong> Metastases<strong>and</strong> Immunohistochemical Features . . . 2769.6.6 Differential Diagnosis . . . . . . . . . . . . 2769.6.7 Treatment <strong>and</strong> Results . . . . . . . . . . . . 2789.7 <strong>Neck</strong> Dissection . . . . . . . . . . . . . . . 2789.7.1 Classification <strong>of</strong> <strong>Neck</strong> Dissections . . . . . 2789.7.2 Gross Examination<strong>of</strong> <strong>Neck</strong> Dissection Surgical Specimens . . 2799.7.3 Histologic Evaluation<strong>of</strong> <strong>Neck</strong> Dissection . . . . . . . . . . . . . . 279References . . . . . . . . . . . . . . . . . . 28010 Eye <strong>and</strong> Ocular Adnexa . . . . . . . . . 282M.R. Canninga-Van Dijk10.1 Summary <strong>of</strong> Anatomy<strong>and</strong> Histology . . . . . . . . . . . . . . . . 28410.1.1 Conjunctiva . . . . . . . . . . . . . . . . . 28410.1.2 Cornea . . . . . . . . . . . . . . . . . . . . 28410.1.3 Intraocular Tissues . . . . . . . . . . . . . 28410.1.4 Optic Nerve . . . . . . . . . . . . . . . . . 28410.1.5 Lacrimal Gl<strong>and</strong>s<strong>and</strong> Lacrimal Passages . . . . . . . . . . . 28410.1.6 Eyelids . . . . . . . . . . . . . . . . . . . . . 28410.1.7 Orbit . . . . . . . . . . . . . . . . . . . . . . 28510.2 Conjunctiva . . . . . . . . . . . . . . . . . . 28510.2.1 Developmental Anomalies . . . . . . . . . 28510.2.1.1 Dermoid, Dermolipoma<strong>and</strong> Complex Choristoma . . . . . . . . . 28510.2.2 Cysts . . . . . . . . . . . . . . . . . . . . . . 28510.2.2.1 Inclusion cysts . . . . . . . . . . . . . . . . 28510.2.3 Degeneration . . . . . . . . . . . . . . . . . 28610.2.3.1 Pinguecula <strong>and</strong> Pterygium . . . . . . . . . 28610.2.4 Inflammatory Processes . . . . . . . . . . 28610.2.4.1 Acute Conjunctivitis . . . . . . . . . . . . 28610.2.4.2 Chronic Non-GranulomatousConjunctivitis . . . . . . . . . . . . . . . . 28710.2.4.3 Granulomatous Conjunctivitis . . . . . . 28710.2.4.4 Ligneous Conjunctivitis . . . . . . . . . . 28710.2.4.5 Chlamydia Trachomatis (TRIC Agent)Infection . . . . . . . . . . . . . . . . . . . 28710.2.5 Dermatologic<strong>and</strong> Systemic Diseases . . . . . . . . . . . 28810.2.5.1 Keratoconjunctivitis Sicca . . . . . . . . . 28810.2.5.2 Dermatologic Diseases . . . . . . . . . . . 28810.2.5.3 Metabolic Diseases . . . . . . . . . . . . . 28810.2.6 Tumours<strong>and</strong> Tumour-Like Conditions . . . . . . . 28810.2.6.1 Epi<strong>the</strong>lial . . . . . . . . . . . . . . . . . . . 28810.2.6.2 Melanocytic . . . . . . . . . . . . . . . . . 29010.2.6.3 O<strong>the</strong>r Neoplasms . . . . . . . . . . . . . . 29110.3 Cornea . . . . . . . . . . . . . . . . . . . . 29210.3.1 Keratitis <strong>and</strong> Corneal Ulcers . . . . . . . . 29210.3.1.1 Herpes Simplex Keratitis . . . . . . . . . . 29210.3.1.2 Corneal UlcerationDue to Systemic Disease . . . . . . . . . . 29210.3.2 Keratoconus . . . . . . . . . . . . . . . . . 29210.3.3 Hereditary Corneal Dystrophies . . . . . 29310.3.3.1 Epi<strong>the</strong>lial Dystrophies . . . . . . . . . . . 29310.3.3.2 Stromal Dystrophies . . . . . . . . . . . . 29310.3.3.3 Endo<strong>the</strong>lial Dystrophies . . . . . . . . . . 29310.3.4 Failed Previous Grafts . . . . . . . . . . . . 294


ContentsXIX10.4 Intraocular Tissues . . . . . . . . . . . . . 29410.4.1 Developmental Anomalies . . . . . . . . . 29410.4.1.1 Congenital Glaucoma . . . . . . . . . . . . 29410.4.1.2 Retinopathy <strong>of</strong> Prematurity . . . . . . . . 29410.4.1.3 Persistent Primary HyperplasticVitreous . . . . . . . . . . . . . . . . . . . . 29410.4.1.4 Retinal Dysplasia . . . . . . . . . . . . . . . 29410.4.1.5 Aniridia . . . . . . . . . . . . . . . . . . . . 29410.4.1.6 Congenital Rubella Syndrome . . . . . . . 29410.4.2 Inflammatory Processes . . . . . . . . . . 29510.4.2.1 Acute Inflammation . . . . . . . . . . . . . 29510.4.2.2 Chronic Non-GranulomatousInflammation . . . . . . . . . . . . . . . . 29510.4.2.3 Granulomatous Inflammation . . . . . . . 29510.4.3 Trauma . . . . . . . . . . . . . . . . . . . . 29610.4.4 Degeneration . . . . . . . . . . . . . . . . . 29610.4.4.1 Glaucoma . . . . . . . . . . . . . . . . . . . 29610.4.4.2 Cataracts . . . . . . . . . . . . . . . . . . . 29710.4.4.3 Phtisis Bulbi . . . . . . . . . . . . . . . . . 29810.4.4.4 Retinal Vascular Disease . . . . . . . . . . 29810.4.4.5 Retinal Detachment . . . . . . . . . . . . . 29810.4.4.6 Retinitis Pigmentosa . . . . . . . . . . . . . 29810.4.5 Tumours<strong>and</strong> Tumour-Like Conditions . . . . . . . 29810.4.5.1 Melanocytic . . . . . . . . . . . . . . . . . 29810.4.5.2 Lymphoid . . . . . . . . . . . . . . . . . . . 30010.4.5.3 Retinoblastoma<strong>and</strong> Pseudoretinoblastoma . . . . . . . . . 30010.4.5.4 Glial . . . . . . . . . . . . . . . . . . . . . . 30110.4.5.5 Vascular . . . . . . . . . . . . . . . . . . . . 30110.4.5.6 O<strong>the</strong>r Primary Tumours . . . . . . . . . . 30110.4.5.7 Metastatic Tumours . . . . . . . . . . . . . 30210.5 Optic Nerve . . . . . . . . . . . . . . . . . 30210.5.1 Papilloedema . . . . . . . . . . . . . . . . . 30210.5.2 Optic Neuritis . . . . . . . . . . . . . . . . 30210.5.3 Optic Atrophy . . . . . . . . . . . . . . . . 30210.5.4 Tumours . . . . . . . . . . . . . . . . . . . 30210.5.4.1 Glioma . . . . . . . . . . . . . . . . . . . . 30210.5.4.2 Meningioma . . . . . . . . . . . . . . . . . 30210.6 Lacrimal Gl<strong>and</strong><strong>and</strong> Lacrimal Passages . . . . . . . . . . . 30210.6.1 Inflammatory Processes . . . . . . . . . . 30210.6.2 Tumours<strong>and</strong> Tumour-Like Conditions . . . . . . . 30310.7 Eyelids . . . . . . . . . . . . . . . . . . . . . 30310.7.1 Cysts . . . . . . . . . . . . . . . . . . . . . . 30310.7.1.1 Dermoid Cyst . . . . . . . . . . . . . . . . 30310.7.1.2 Epidermal Cyst . . . . . . . . . . . . . . . 30310.7.1.3 Hidrocystoma . . . . . . . . . . . . . . . . 30310.7.2 Inflammatory Processes . . . . . . . . . . 30310.7.2.1 Chalazion <strong>and</strong> O<strong>the</strong>r Ruptured Cysts . . . 30410.7.2.2 Deep Granuloma Annulare . . . . . . . . . 30410.7.2.3 Necrobiotic Xanthogranuloma . . . . . . . 30410.7.3 Amyloidosis . . . . . . . . . . . . . . . . . 30510.7.4 Tumours<strong>and</strong> Tumour-Like Conditions . . . . . . . 30510.7.4.1 Xan<strong>the</strong>lasmata . . . . . . . . . . . . . . . . 30510.8 Orbit . . . . . . . . . . . . . . . . . . . . . . 30510.8.1 Inflammatory Processes . . . . . . . . . . 30510.8.1.1 Dysthyroid Ophthalmopathy . . . . . . . 30510.8.1.2 Cellulitis . . . . . . . . . . . . . . . . . . . 30510.8.1.3 Pseudotumour . . . . . . . . . . . . . . . . 30610.8.2 Tumours<strong>and</strong> Tumour-Like Conditions . . . . . . . 30610.8.2.1 Developmental Cysts . . . . . . . . . . . . 30610.8.2.2 Optic Nerve<strong>and</strong> Meningeal Tumours . . . . . . . . . . 30610.8.2.3 Metastatic Tumours . . . . . . . . . . . . . 307References . . . . . . . . . . . . . . . . . . 307Subject Index . . . . . . . . . . . . . . . 311


List <strong>of</strong> ContributorsLlucia Alos(e-mail: lalos@clinic.ub.es)Department <strong>of</strong> Pathological Anatomy,Hospital Clinic, University <strong>of</strong> Barcelona,Villarroel 170, 08036 Barcelona, SpainM.R. Canninga-Van Dijk(e-mail: m.r.canningav<strong>and</strong>ijk@azu.nl)Department <strong>of</strong> <strong>Pathology</strong>,University Medical Centre Utrecht, H04-312,P.O. Box 85500, 3508 GA, Utrecht, The Ne<strong>the</strong>rl<strong>and</strong>sAntonio Cardesa(e-mail: acardesa@clinic.ub.es)Department <strong>of</strong> Pathological Anatomy,Hospital Clinic, University <strong>of</strong> Barcelona,Villarroel 170, 08036 Barcelona, SpainSilvana Di Palma(e-mail: silvana.dipalma@royalsurrey.nhs.uk)Department <strong>of</strong> Histopathology,University <strong>of</strong> Surrey, Royal Surrey County Hospital,Egerton Road, Guildford, GU2 7XX, UKJohn Wallace Eveson(e-mail: j.w.eveson@bristol.ac.uk)Division <strong>of</strong> Oral Medicine,<strong>Pathology</strong> <strong>and</strong> Microbiology,University <strong>of</strong> Bristol Dental School,Lower Maudlin Street, Bristol, BS1 2LY, UKAless<strong>and</strong>ro Franchi(e-mail: franchi@unifi.it)Department <strong>of</strong> Human <strong>Pathology</strong> <strong>and</strong> Oncology,University <strong>of</strong> Florence,Viale Morgagni 85, 50134 Florence, ItalyNina Gale(e-mail: nina.gale@mf.uni-lj.si)Institute <strong>of</strong> <strong>Pathology</strong>, Faculty <strong>of</strong> Medicine,University <strong>of</strong> Ljubljana,Korytkova 2, 1000 Ljubljana, SloveniaIlmo Leivo(e-mail: ilmo.leivo@helsinki.fi)Department <strong>of</strong> <strong>Pathology</strong>, Haartman Institute,P.O. Box 21 (Haartmaninkatu 3)00014 University <strong>of</strong> Helsinki, Helsinki, Finl<strong>and</strong>Mario A. Luna(e-mail: mluna@md<strong>and</strong>erson.org)Department <strong>of</strong> <strong>Pathology</strong>, The University <strong>of</strong> Texas,M.D. Anderson Cancer Center,1515 Holcombe Blvd, Box 85,Houston, Texas 77030, USALeslie Michaels(e-mail: l.michaels@ucl.ac.uk)Department <strong>of</strong> Histopathology,Royal Free <strong>and</strong> UCL Medical School,Rockefeller Building,University Street, London WC1E 6JJ, UKKeyla Pineda-DaboinDepartment <strong>of</strong> <strong>Pathology</strong>,Military Hospital “Carlos Arvelo”<strong>and</strong> Institute <strong>of</strong> Anatomical <strong>Pathology</strong>,University Central <strong>of</strong> Venezuela, Caracas, VenezuelaSigrid Regauer(e-mail: sigrid.regauer@meduni-graz.at)Institute <strong>of</strong> <strong>Pathology</strong>,Karl Franzens University <strong>of</strong> Graz,Auenbruggerplatz 25, 8036 Graz, AustriaRoderick H.W. Simpson(e-mail: roderick.simpson@virgin.net)Department <strong>of</strong> Histopathology,Church Lane Exeter, EX2 5AD, UKAlena Skalova(e-mail: skalova@fnplzen.cz)Department <strong>of</strong> <strong>Pathology</strong>,Medical Faculty Hospital,Dr. E Benese 13, 305 99 Plzen, Czech Republic


XXIIList <strong>of</strong> ContributorsPieter J. Slootweg(e-mail: p.slootweg@pathol.umcn.nl)Department <strong>of</strong> <strong>Pathology</strong>,University Medical Center St. Radboud, HP 437,P.O. Box 9101, 6500 HB Nijmegen, The Ne<strong>the</strong>rl<strong>and</strong>sNina Zidar(e-mail: nina.zidar@mf.uni-lj.si)Institute <strong>of</strong> <strong>Pathology</strong>, Faculty <strong>of</strong> Medicine,University <strong>of</strong> Ljubljana,Korytkova 2, 1000 Ljubljana, Slovenia


Chapter 1Benign <strong>and</strong> Potentially Malignant Lesions<strong>of</strong> <strong>the</strong> Squamous Epi<strong>the</strong>lium<strong>and</strong> Squamous Cell CarcinomaN. Gale · N. Zidar1Contents1.1 Squamous Cell Papilloma <strong>and</strong> Related Lesions . . . . . 21.1.1 Squamous Cell Papilloma,Verruca Vulgaris, Condyloma Acuminatum<strong>and</strong> Focal Epi<strong>the</strong>lial Hyperplasia . . . . . . . . . . . . 21.1.2 Laryngeal Papillomatosis . . . . . . . . . . . . . . . . . 31.2 Squamous Intraepi<strong>the</strong>lial Lesions (SILS) . . . . . . . . 41.2.1 General Considerations . . . . . . . . . . . . . . . . . . 41.2.2 Terminological Problems . . . . . . . . . . . . . . . . . 41.2.3 Aetiology . . . . . . . . . . . . . . . . . . . . . . . . . . . 51.2.3.1 Oral Cavity <strong>and</strong> Oropharyn . . . . . . . . . . . . . . . . 51.2.3.2 Larynx . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51.2.4 Clinical Features<strong>and</strong> Macroscopic Appearances . . . . . . . . . . . . . . 61.2.4.1 Oral <strong>and</strong> Oropharyngeal Leukoplakia, ProliferativeVerrucous Leukoplakia <strong>and</strong> Erythroplakia . . . . . . . 61.2.4.2 Laryngeal <strong>and</strong> Hypopharyngeal Leukoplakia<strong>and</strong> Chronic Laryngitis . . . . . . . . . . . . . . . . . . 71.2.5 Histological Classifications . . . . . . . . . . . . . . . . 81.2.5.1 WHO Dysplasia System . . . . . . . . . . . . . . . . . . 81.2.5.2 The Ljubljana Classification . . . . . . . . . . . . . . . . 91.2.5.3 Comparison Between <strong>the</strong> Ljubljana Classification<strong>and</strong> WHO 2005 Classification . . . . . . . . . . . . . . . 111.2.6 Biomarkers Related to Malignant Potential <strong>of</strong> SILsRecognised by Auxiliary <strong>and</strong> Advanced MolecularMethods . . . . . . . . . . . . . . . . . . . . . . . . . . . 121.2.7 Treatment <strong>and</strong> Prognosis . . . . . . . . . . . . . . . . . 121.2.7.1 Oral Cavity <strong>and</strong> Oropharynx . . . . . . . . . . . . . . . 121.2.7.2 Larynx . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131.3 Invasive Squamous Cell Carcinoma . . . . . . . . . . . 131.3.1 Microinvasive Squamous Cell Carcinoma . . . . . . . 131.3.2 Conventional Squamous Cell Carcinoma . . . . . . . . 131.3.2.1 Aetiology . . . . . . . . . . . . . . . . . . . . . . . . . . . 141.3.2.2 Pathologic Features . . . . . . . . . . . . . . . . . . . . . 141.3.2.3 Grading . . . . . . . . . . . . . . . . . . . . . . . . . . . . 141.3.2.4 Invasive Front . . . . . . . . . . . . . . . . . . . . . . . . 151.3.2.5 Stromal Reaction . . . . . . . . . . . . . . . . . . . . . . 151.3.2.6 Differential Diagnosis . . . . . . . . . . . . . . . . . . . 151.3.2.7 Treatment <strong>and</strong> Prognosis . . . . . . . . . . . . . . . . . 151.3.3 Spindle Cell Carcinoma . . . . . . . . . . . . . . . . . . 161.3.3.1 Aetiology . . . . . . . . . . . . . . . . . . . . . . . . . . . 161.3.3.2 Pathologic Features . . . . . . . . . . . . . . . . . . . . . 161.3.3.3 Differential Diagnosis . . . . . . . . . . . . . . . . . . . 171.3.3.4 Treatment <strong>and</strong> Prognosis . . . . . . . . . . . . . . . . . 171.3.4 Verrucous Carcinoma . . . . . . . . . . . . . . . . . . . 171.3.4.1 Aetiology . . . . . . . . . . . . . . . . . . . . . . . . . . . 171.3.4.2 Pathologic Features . . . . . . . . . . . . . . . . . . . . . 181.3.4.3 Differential Diagnosis . . . . . . . . . . . . . . . . . . 181.3.4.4 Treatment . . . . . . . . . . . . . . . . . . . . . . . . . 181.3.4.5 Prognosis . . . . . . . . . . . . . . . . . . . . . . . . . . 191.3.5 Papillary Squamous Cell Carcinoma . . . . . . . . . . 191.3.5.1 Aetiology . . . . . . . . . . . . . . . . . . . . . . . . . . 191.3.5.2 Pathologic Features . . . . . . . . . . . . . . . . . . . . 191.3.5.3 Differential Diagnosis . . . . . . . . . . . . . . . . . . 201.3.5.4 Treatment <strong>and</strong> Prognosis . . . . . . . . . . . . . . . . 201.3.6 Basaloid Squamous Cell Carcinoma . . . . . . . . . . 201.3.6.1 Aetiology . . . . . . . . . . . . . . . . . . . . . . . . . . 201.3.6.2 Pathologic Features . . . . . . . . . . . . . . . . . . . . 201.3.6.3 Differential Diagnosis . . . . . . . . . . . . . . . . . . 211.3.6.4 Treatment <strong>and</strong> Prognosis . . . . . . . . . . . . . . . . 211.3.7 Adenoid Squamous Cell Carcinoma . . . . . . . . . . 221.3.7.1 Pathologic Features . . . . . . . . . . . . . . . . . . . . 221.3.7.2 Differential Diagnosis . . . . . . . . . . . . . . . . . . 221.3.7.3 Treatment <strong>and</strong> Prognosis . . . . . . . . . . . . . . . . 221.3.8 Adenosquamous Carcinoma . . . . . . . . . . . . . . 231.3.8.1 Aetiology . . . . . . . . . . . . . . . . . . . . . . . . . . 231.3.8.2 Pathologic Features . . . . . . . . . . . . . . . . . . . . 231.3.8.3 Differential Diagnosis . . . . . . . . . . . . . . . . . . 231.3.8.4 Treatment <strong>and</strong> Prognosis . . . . . . . . . . . . . . . . 241.3.9 Lymphoepi<strong>the</strong>lial Carcinoma . . . . . . . . . . . . . . 241.3.9.1 Aetiology . . . . . . . . . . . . . . . . . . . . . . . . . . 241.3.9.2 Pathologic Features . . . . . . . . . . . . . . . . . . . . 241.3.9.3 Differential Diagnosis . . . . . . . . . . . . . . . . . . 251.3.9.4 Treatment <strong>and</strong> Prognosis . . . . . . . . . . . . . . . . 251.4 Second Primary Tumours . . . . . . . . . . . . . . . . 251.5 Tumour Spread <strong>and</strong> Metastasising . . . . . . . . . . . 251.5.1 Invasion <strong>of</strong> Lymphatic <strong>and</strong> Blood Vessels . . . . . . . 261.5.2 Perineural Invasion . . . . . . . . . . . . . . . . . . . . 261.5.3 Regional Lymph Node Metastases . . . . . . . . . . . 261.5.3.1 Extracapsular Spreadin Lymph Node Metastases . . . . . . . . . . . . . . . 261.5.3.2 Metastases in <strong>the</strong> S<strong>of</strong>t Tissue <strong>of</strong> <strong>the</strong> <strong>Neck</strong> . . . . . . . 271.5.4 Distant Metastasis . . . . . . . . . . . . . . . . . . . . 271.5.5 Micrometastasis . . . . . . . . . . . . . . . . . . . . . . 271.6 Molecular <strong>Pathology</strong><strong>of</strong> Squamous Cell Carcinoma . . . . . . . . . . . . . . 281.6.1 Detecting Tumour Cells . . . . . . . . . . . . . . . . . 281.6.2 Clonal Analysis . . . . . . . . . . . . . . . . . . . . . . 281.6.3 Assessment <strong>of</strong> Risk for Malignant Progression . . . . 291.6.4 DNA/RNA Pr<strong>of</strong>ilingin Predicting Metastatic Disease . . . . . . . . . . . . 29References . . . . . . . . . . . . . . . . . . . . . . . . . 29


2 N. Gale · N. Zidar11.1 Squamous Cell Papilloma<strong>and</strong> Related LesionsBenign, exophytic, papillary or verrucous lesions <strong>of</strong> <strong>the</strong>squamous epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong> oral cavity, oropharynx<strong>and</strong> larynx include similar entities such as squamouscell papilloma (SCP), verruca vulgaris (VV), condylomaacuminatum (CA), <strong>and</strong> focal epi<strong>the</strong>lial hyperplasia(FEH). However, not every papillary lesion in <strong>the</strong>seareas can be placed into one <strong>of</strong> <strong>the</strong> listed categories. Itseems that <strong>the</strong> majority <strong>of</strong> lesions are similar variants <strong>of</strong>mucosal proliferations, frequently induced by infectionsby human papillomaviruses (HPV). They show more orless overlapping clinical <strong>and</strong> morphological properties,but different biological behaviour, ranging from ra<strong>the</strong>rinconspicuous to potentially life threatening. Classification<strong>of</strong> <strong>the</strong>se changes into infectious (VV, CA, FEH), <strong>and</strong>neoplastic (SCP), is thought to be ra<strong>the</strong>r inconsistent <strong>and</strong>not well founded. Papillary lesions, except for laryngealpapillomatosis, generally have a favourable outcome.1.1.1 Squamous Cell Papilloma,Verruca Vulgaris,Condyloma Acuminatum<strong>and</strong> Focal Epi<strong>the</strong>lial HyperplasiaICD-O:8052/0Squamous cell papilloma, <strong>the</strong> most frequent papillarylesion <strong>of</strong> <strong>the</strong> oral cavity <strong>and</strong> oropharynx, is usually asingle, pedunculated, white or pink lesion, consisting <strong>of</strong>finger-like mucosal projections (Fig. 1.1). It may occasionallybe sessile with a granular or verrucous surface.The lesion, usually smaller than 1 cm, grows rapidly<strong>and</strong> has predilections for <strong>the</strong> hard <strong>and</strong> s<strong>of</strong>t palate <strong>and</strong>lateral border <strong>of</strong> <strong>the</strong> tongue [2, 285]. Multiple sessile lesionsin children are characteristic <strong>of</strong> VV; <strong>the</strong>y are foundon <strong>the</strong> lips, palate <strong>and</strong> gingiva. CAs are usually largerthan SCPs, multiple dome-shaped nodular lesions thatmainly appear on <strong>the</strong> lips <strong>and</strong> s<strong>of</strong>t palate. FEHs are characterisedby multiple sessile or elevated papules, usuallydistributed over <strong>the</strong> buccal, labial <strong>and</strong> tongue mucosa.Aetiologically, it is extremely difficult to establish<strong>the</strong>ir accurate relationship to HPV infection due to variationsin tissue samplings, <strong>the</strong> ethnic <strong>and</strong> geographic origin<strong>of</strong> patients, <strong>and</strong> <strong>the</strong> use <strong>of</strong> non-molecular vs. molecularmethods for HPV detection with different levels<strong>of</strong> sensitivity [285, 374]. However, more than 20 HPVgenotypes have been detected in oral papillary lesions[285]. SCPs are mainly related to HPV genotypes 6 <strong>and</strong>11 [386], VV to HPV genotypes 2, 4, 6, 11, <strong>and</strong> 16 [142,244], CA to HPV genotypes 6, 11, 16, <strong>and</strong> 18 [100, 201]<strong>and</strong> FEH to HPV genotypes 13 <strong>and</strong> 32 [285, 286]. Onlya few cases <strong>of</strong> VV have been described in <strong>the</strong> larynx.Barnes <strong>and</strong> co-workers studied a single case <strong>and</strong> unexpectedlyfound it to contain HPV genotypes 6 <strong>and</strong> 11 <strong>and</strong>not genotypes 2 <strong>and</strong> 4, which are characteristic <strong>of</strong> mucosalVV [22]. O<strong>the</strong>r, non-infectious aetiological factorsare not well known for oral papillary lesions (Fig. 1.1).Histologically, SCPs are composed <strong>of</strong> narrow papillaryprojections <strong>of</strong> s<strong>of</strong>t fibrous stroma covered by keratoticor parakeratotic squamous epi<strong>the</strong>lium (Fig. 1.2).Koilocytosis, <strong>the</strong> only visible cytopathic effect <strong>of</strong>HPV infection, which is caused by viral replication in<strong>the</strong> upper intermediate <strong>and</strong> superficial zone <strong>of</strong> <strong>the</strong> squamousepi<strong>the</strong>lium, is rarely visible in SCPs. VV showssimilar histological features, but peripheral papillaryprojections are usually centrally bend, <strong>and</strong> koilocytosis<strong>and</strong> <strong>the</strong> granular layer are prominent. The characteristics<strong>of</strong> CAs are obvious: koilocytosis <strong>and</strong> bulbous reteridges <strong>of</strong> <strong>the</strong> covering epi<strong>the</strong>lium [100, 285]. Koilocytosis,apoptotic bodies <strong>and</strong> epi<strong>the</strong>lial hyperplasia are significantin FEH [61, 285].In <strong>the</strong> differential diagnosis <strong>of</strong> squamous cell oralpapillary lesions, verrucous carcinoma is <strong>the</strong> most importantconsideration. An evident downgrowth <strong>of</strong> bulbousepi<strong>the</strong>lial projections favours a diagnosis <strong>of</strong> verrucouscarcinoma. Oral SCPs in patients with acquired im-Fig. 1.1. Whitish papillary lesion <strong>of</strong> <strong>the</strong> palate. Courtesy <strong>of</strong> Dr. J.Fischinger, Ljubljana, SloveniaFig. 1.2. Oral squamous cell papilloma. Projections <strong>of</strong> fibrovascularstroma are covered by parakeratotic squamous epi<strong>the</strong>lium


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 3munodeficiency syndrome (AIDS) may show a certainamount <strong>of</strong> epi<strong>the</strong>lial atypia. In <strong>the</strong>se cases SCPs have tobe differentiated from squamous cell carcinoma [295].The treatment for SCPs <strong>and</strong> related papillary lesionsis surgical removal. The infectivity <strong>of</strong> HPV in SCPs isvery low <strong>and</strong> recurrence uncommon, except in lesionsassociated with human immunodeficiency virus (HIV)infections. On <strong>the</strong> o<strong>the</strong>r h<strong>and</strong>, recurrence is more commonin CAs. No special treatment is required for FEHunless <strong>the</strong> lesions are extensive.1.1.2 Laryngeal PapillomatosisFig. 1.3. Laryngeal papillomatosis. Numerous clusters <strong>of</strong> papillomasobliterate <strong>the</strong> laryngeal lumenICD-O:8060/0Laryngeal squamous cell papillomas (LSCPs) are <strong>the</strong>most frustrating benign lesions in <strong>the</strong> head <strong>and</strong> neckregion. Because <strong>of</strong> <strong>the</strong>ir clinical specificities, such asmultiplicity, recurrence <strong>and</strong> <strong>the</strong> propensity to spread toadjacent areas, it has been suggested that LSCPs shouldbe renamed recurrent respiratory papillomatosis (RRP)[34, 89, 91, 187].Recurrent respiratory papillomatosis is aetiologicallyrelated to HPV [4, 212, 283, 289, 352]. HPV-6 <strong>and</strong> -11 are <strong>the</strong> most frequent genotypes associated with RRP(Fig. 1.4b) [4, 126, 212, 284, 289, 330].Characteristically, LSCPs show a bimodal age distribution:<strong>the</strong> first peak is before <strong>the</strong> age <strong>of</strong> 5 years with nogender predominance; <strong>the</strong> second peak occurs between<strong>the</strong> ages <strong>of</strong> 20 <strong>and</strong> 40 years with a male to female ratio <strong>of</strong>3:2 [34, 87, 91, 189, 216].Human papillomavirus transmission in children isassociated with perinatal transmission from an infectedmo<strong>the</strong>r to <strong>the</strong> child [34, 88, 217]. The mode <strong>of</strong> HPVinfection in adults remains unclear. The reactivation<strong>of</strong> a latent infection acquired perinatally or a postpartuminfection with orogenital contacts has been suggested[4, 188]. In contrast to RRP, a solitary keratinisingsquamous papilloma or papillary keratosis <strong>of</strong> adults appearsnot to be associated with viral infection, althoughit may recur or be occasionally associated with malignanttransformation [20].Recurrent respiratory papillomatosis almost invariablyinvolves <strong>the</strong> larynx, especially <strong>the</strong> true <strong>and</strong> false vocalcords, subglottic areas <strong>and</strong> ventricles [4]. An extralaryngealspread may occur successively to <strong>the</strong> oral cavity,trachea <strong>and</strong> bronchi. Although RRP has been traditionallydivided into juvenile <strong>and</strong> adult groups [87, 189,216, 352], <strong>the</strong> prevailing opinion has recognised <strong>the</strong> diseaseas a unified biological entity with differences inclinical courses, caused by HPV genotypes 6 or 11 [28,126, 189, 218, 321]. For children, multiple <strong>and</strong> extensivegrowth with rapid recurrence after excision is characteristic.The small diameter <strong>of</strong> <strong>the</strong> airways in childrenmay cause dangerous or even fatal airway obstruction.The clinical course in adults is usually not so dramatic,although RRP can be aggressive with multiple recurrences[43, 284]. Most children present with dysphonia<strong>and</strong> stridor, <strong>and</strong> less commonly with a chronic cough,recurrent pneumonia, dyspnoea, <strong>and</strong> acute life-threateningevents [34, 43, 88]. Affected adults present mostlywith dysphonia <strong>and</strong> hoarseness [43, 181].Grossly, papillomas are exophytic, branching, pedunculateor sessile masses, pink or reddish in colour, witha finely lobulated surface, presenting ei<strong>the</strong>r singly or inclusters (Fig. 1.3).Histologically, RRP is composed <strong>of</strong> finger-like projections<strong>of</strong> <strong>the</strong> squamous epi<strong>the</strong>lium, covering thin fibrovascularcores. A basal <strong>and</strong> parabasal hyperplasia <strong>of</strong><strong>the</strong> squamous epi<strong>the</strong>lium is most frequently seen, usuallyextending up to <strong>the</strong> mid-portion (Fig. 1.4a). Mitoticfeatures may be prominent within this area. Irregularlyscattered clusters <strong>of</strong> koilocytes are seen in <strong>the</strong> upperpart <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium. Epi<strong>the</strong>lial changes, such as mildto moderate nuclear atypia <strong>and</strong> hyperchromatism, increasednuclear cytoplasmic ratio, increased mitotic activitywith pathological features, <strong>and</strong> prominent surfacekeratinisation are rarely found in RRP [181].Various lesions with a papillary structure must beconsidered in <strong>the</strong> differential diagnosis <strong>of</strong> RRP. In verrucouscarcinomas, <strong>the</strong> squamous fronds are thicker <strong>and</strong>are covered by a prominent keratotic layer, bulbous retepegs infiltrate fibrous stroma in a blunt, pushing manner<strong>and</strong> koilocytosis is usually absent. The papillary squamouscarcinoma usually shows an architectonic similarityto RRP. In contrast to RRP, papillary structures in <strong>the</strong>papillary squamous carcinoma are covered by a clearlyneoplastic epi<strong>the</strong>lium showing invasive growth.The clinical course <strong>of</strong> RRP is unpredictable, characterisedby periods <strong>of</strong> active disease <strong>and</strong> remissions. HPVpresent in apparently normal mucosa serves as a virusreservoir responsible for repeated recurrence <strong>of</strong> papillo-


4 N. Gale · N. Zidar1An exact <strong>and</strong> uniform terminology <strong>of</strong> SILs is a prerequisitefor successful cooperation among pathologists aswell as adequate underst<strong>and</strong>ing with clinicians. A considerableoverlapping <strong>of</strong> clinical <strong>and</strong> histological termsrelating to SILs has been widely noticed due to inadeaFig. 1.4. Laryngeal papillomatosis. a Branches <strong>of</strong> laryngeal papillomaare covered with hyperplastic squamous cell epi<strong>the</strong>lium. Numerouskoilocytes are seen in <strong>the</strong> upper part <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium.bb Positive in situ hybridisation signal for HPV genotypes 6 <strong>and</strong> 11in an adult laryngeal papillomamas [301, 330]. The presence <strong>of</strong> RRP in <strong>the</strong> neonatal periodis a negative prognostic factor with a greater needfor tracheotomy <strong>and</strong> likelihood <strong>of</strong> mortality [88]. Onereport on <strong>the</strong> spontaneous disappearance <strong>of</strong> <strong>the</strong> disease,especially during puberty, has not been fur<strong>the</strong>r supported[4]. Increased histologic changes (atypia <strong>of</strong> epi<strong>the</strong>lialcells) are reported to be associated with increased severity<strong>and</strong> recurrence <strong>of</strong> RRP [75, 288]. O<strong>the</strong>rs have suggestedthat <strong>the</strong> histologic changes <strong>of</strong> RRP are not a goodpredictor <strong>of</strong> eventual malignant transformation [133].Malignant transformation occurs mainly in patientswith a history <strong>of</strong> previous irradiation or heavy smoking[290], <strong>and</strong> rarely without any predisposing factors[143, 296]. In children, carcinomas preferentially appearin <strong>the</strong> bronchopulmonary tree, <strong>and</strong> in adults in <strong>the</strong> larynx[141]. HPV genotype 11 is assumed to be most frequentlyassociated with malignant transformation <strong>of</strong>RRP [70, 206, 218, 290], followed by HPV-16 [92] <strong>and</strong>HPV-18 [311].The overall mortality rate <strong>of</strong> patients with RRP rangesfrom 4 to 14% [20], <strong>and</strong> is mostly causally related toasphyxia, pulmonary complications <strong>and</strong> cancer development[17, 20, 338].1.2 Squamous Intraepi<strong>the</strong>lial Lesionspression <strong>of</strong> <strong>the</strong> whole spectrum <strong>of</strong> epi<strong>the</strong>lial changesranging from squamous cell hyperplasia to carcinomain situ.It has been widely accepted that <strong>the</strong> transition fromnormal mucosa to invasive squamous cell carcinoma(SCC) is a comprehensive <strong>and</strong> multistage process, causallyrelated to a progressive accumulation <strong>of</strong> geneticchanges leading to <strong>the</strong> selection <strong>of</strong> a clonal population <strong>of</strong>transformed epi<strong>the</strong>lial cells [144]. Between six <strong>and</strong> tenindependent genetic events are required for progressionto SCC [300]. In <strong>the</strong>ir evolution, some cases <strong>of</strong> SIL areself-limiting <strong>and</strong> reversible, some persist, <strong>and</strong> some <strong>of</strong><strong>the</strong>m progress to SCC in spite <strong>of</strong> treatment [78]. Particularinterest has been focused on potentially malignantor risky (precancerous) lesions [48, 181, 200, 223]. Theselesions have been defined as histomorphological changes<strong>of</strong> <strong>the</strong> squamous epi<strong>the</strong>lium from which invasive cancerdevelops in a higher percentage than from o<strong>the</strong>r epi<strong>the</strong>liallesions [125, 179, 181, 223]. A fundamental enigma<strong>of</strong> potentially malignant lesions remains when <strong>and</strong>under what conditions <strong>the</strong>se changes turn to malignantgrowth [180, 223].Various aetiological, clinical, histological <strong>and</strong> moleculargenetic aspects are significant for <strong>the</strong> evaluation,adequate treatment <strong>and</strong> predictive behaviour <strong>of</strong> SILs,particularly <strong>of</strong> potentially malignant lesions.1.2.1 General ConsiderationsHistological changes <strong>of</strong> <strong>the</strong> squamous epi<strong>the</strong>lium thatoccur in <strong>the</strong> process <strong>of</strong> oral, oro- <strong>and</strong> hypopharyngeal<strong>and</strong> laryngeal carcinogenesis, are cumulatively designatedsquamous intraepi<strong>the</strong>lial lesions (SILs). Theterm SILs has been proposed as an all-embracing ex-1.2.2 Terminological Problems


6 N. Gale · N. Zidar1to increase with <strong>the</strong> duration <strong>of</strong> smoking, <strong>the</strong> quality <strong>of</strong>tobacco, <strong>the</strong> practice <strong>of</strong> deep inhalation <strong>and</strong> <strong>the</strong> inabilityto stop smoking, <strong>and</strong> inversely with <strong>the</strong> age <strong>of</strong> <strong>the</strong> patientat <strong>the</strong> start <strong>of</strong> smoking.Additional aetiological factors are: industrial pollution,chronic infections, voice abuse, obstruction <strong>of</strong> <strong>the</strong>upper respiratory tract, vitamin deficiency, <strong>and</strong> hormonaldisturbance [115, 181, 184, 185, 228, 276]. The role <strong>of</strong>HPV infection in laryngeal carcinogenesis remains unclarified[331]. The prevalence <strong>of</strong> HPV infection in laryngealcarcinomas varies significantly among various studies,ranging from 0 to 54.1% [346]. The overall prevalence<strong>of</strong> HPV infection in nine studies <strong>of</strong> SILs [16, 54, 118, 128,136, 137, 219, 281, 302] was found to be 12.4%. However,HPV DNA was also detected in a clinically <strong>and</strong> histologicallynormal larynx in 12–25% <strong>of</strong> individuals [267, 302].Definite evidence <strong>of</strong> an aetiologic role <strong>of</strong> HPV in SIL, atleast at present, is lacking, <strong>and</strong> HPV infection in SILs mayrepresent an incidental HPV colonisation ra<strong>the</strong>r than trueinfection <strong>of</strong> <strong>the</strong> laryngeal mucosa.1.2.4 Clinical Features<strong>and</strong> Macroscopic Appearances1.2.4.1 Oral<strong>and</strong> Oropharyngeal Leukoplakia,Proliferative VerrucousLeukoplakia <strong>and</strong> ErythroplakiaBoth oral leukoplakia (OL) <strong>and</strong> oral erythroplakia (OE)have generally been defined as premalignant lesions,mainly on <strong>the</strong> basis <strong>of</strong> <strong>the</strong>ir clinical appearance [14, 371].It seems more reasonable to disregard clinically basedpremalignant connotations, especially for OL, withoutknowing <strong>the</strong> histological features [200, 297, 342]. Therisk <strong>of</strong> OL becoming malignant is relatively low <strong>and</strong>quite unpredictable [342]. In contrast, OE is a muchmore worrisome lesion than OL <strong>and</strong> always requireshistological evaluation.Oral leukoplakia is a clinical diagnosis <strong>of</strong> exclusion.If any oral white patch can be diagnosed as some o<strong>the</strong>rcondition, such as c<strong>and</strong>idiasis, leukoedema, whitesponge naevus, lichen planus, frictional keratosis, nicotinestomatitis, etc. <strong>the</strong>n <strong>the</strong> lesion should not be considereda case <strong>of</strong> OL [263]. The white appearance <strong>of</strong> OLis most <strong>of</strong>ten related to an increase in <strong>the</strong> surface keratinlayer. OL affects approximately 3% <strong>of</strong> white adults[46]. It is most frequently seen in middle-aged <strong>and</strong> oldermen with an increasing prevalence with age, reaching8% in men over 70 years [48, 49]. However, recent studiesreported a tendency towards a lower prevalence <strong>of</strong>OL, compared with <strong>the</strong> past, which might be <strong>the</strong> result<strong>of</strong> <strong>the</strong> massive public health education campaign againsttobacco [314].Fig. 1.5. Leukoplakia <strong>of</strong> <strong>the</strong> dorsal tongue. The microscopicdiagnosis was basal <strong>and</strong> parabasal cell hyperplasia. Courtesy<strong>of</strong> Dr. J. Fischinger, Ljubljana, SloveniaThe most common sites <strong>of</strong> lesions are <strong>the</strong> buccal <strong>and</strong>alveolar mucosa <strong>and</strong> <strong>the</strong> lower lip. Lesions in <strong>the</strong> floor <strong>of</strong><strong>the</strong> mouth, lateral tongue <strong>and</strong> lower lip more <strong>of</strong>ten showepi<strong>the</strong>lial atypia or even malignant growth [263]. A consensushas been attained to divide OL clinically into homogenous<strong>and</strong> non-homogenous types [14]. The formertype is characterised as a uniform, flat, thin lesion with asmooth or wrinkled surface showing shallow cracks, buta constant texture throughout (Fig 1.5). The latter typeis defined as a predominantly white or white <strong>and</strong> red lesionthat may be irregularly flat, nodular or exophytic.Nodular lesions have slightly raised rounded, red <strong>and</strong>/orwhitish excrescences. Exophytic lesions have irregularblunt or sharp projections [14]. The term non-homogenousis applicable to <strong>the</strong> aspect <strong>of</strong> both colour (a mixedwhite <strong>and</strong> red lesion) <strong>and</strong> texture (exophytic, papillaryor verrucous) <strong>of</strong> <strong>the</strong> lesions (Fig. 1.6).With regard to verrucous lesions, <strong>the</strong>re are no reproducibleclinical criteria to distinguish among verrucoushyperplasia, proliferative verrucous hyperplasia <strong>and</strong>verrucous carcinoma [371]. Any persisting lesion withno apparent aetiology should be considered suspicious[235]. A period <strong>of</strong> 2–4 weeks seems acceptable to observe<strong>the</strong> regression or disappearance <strong>of</strong> <strong>the</strong> OL after <strong>the</strong>elimination <strong>of</strong> possible causative factors. After that timea biopsy is obligatory [371].Proliferative verrucous hyperplasia (PVL) is a specialtype <strong>of</strong> OL with a proven high risk <strong>of</strong> becomingmalignant [32, 322]. Initially, it is relatively benignlooking,a homogenous solitary patch that turns graduallyto an exophytic, diffuse or multifocal, progressive<strong>and</strong> irreversible lesion [32, 322, 390]. The diagnosisis made retrospectively after evidence <strong>of</strong> a progressiveclinical course, accompanied by a particulardeterioration in histological changes. Women predominateover men in PVL by 4 to 1, with a mean age atdiagnosis <strong>of</strong> 62 years [322]. The epidemiology <strong>of</strong> PVL


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 7Fig. 1.6. Erythroleukoplakia <strong>of</strong> <strong>the</strong> buccal mucosa. The microscopicdiagnosis was atypical hyperplasia. Courtesy <strong>of</strong> Dr.D. Dovšak, Ljubljana, Sloveniadoes not highlight a particular causal agent <strong>and</strong> <strong>the</strong> lesionwould appear to be multifactorial [114, 342]. Therelatively common absence <strong>of</strong> well-known risk factorsassociated with oral cancer <strong>and</strong> a preponderance <strong>of</strong> elderlyfemale patients, may indicate a different pathogenesis<strong>of</strong> PVL-related, compared with non-PVL-related,cancer [32]. It appears most frequently in <strong>the</strong> buccalmucosa, followed by <strong>the</strong> gingiva, tongue, <strong>and</strong> floor<strong>of</strong> <strong>the</strong> mouth [322]. The severity <strong>of</strong> histologic featurescorrelates with duration <strong>of</strong> lesion, from benign keratoticlesion to verrucous hyperplasia, <strong>and</strong> finally, upto one <strong>of</strong> three forms <strong>of</strong> SCC: verrucous, conventionalor papillary types [32]. PVL should be considereda possible diagnosis when a specific discrepancy betweenbl<strong>and</strong> histological features <strong>and</strong> aggressive clinicalcourse is established [114]. Whe<strong>the</strong>r verrucous hyperplasiaforms a separate stage in this series <strong>of</strong> histologicalfeatures shown by PVL is debatable, as <strong>the</strong>reseems to be considerable histological overlap betweenthis lesion <strong>and</strong> verrucous carcinoma. Thus, <strong>the</strong>re areno convincing arguments that verrucous hyperplasiais anything o<strong>the</strong>r than a variant <strong>of</strong> verrucous carcinoma[327, 371, 390]. A mean time <strong>of</strong> 7.7 years was foundfrom <strong>the</strong> diagnosis <strong>of</strong> PVL to cancer development in70.3% <strong>of</strong> patients [322]. The treatment <strong>of</strong> PVL continuesto be an unsolved problem with high rates <strong>of</strong> recurrence,since total excision is rarely possible because <strong>of</strong><strong>the</strong> widespread growth [32].Oral erythroplakia is much less common than OL.OE occurs most frequently in older men as a red maculaor plaque with a s<strong>of</strong>t, velvety texture, quite sharplydemarcated <strong>and</strong> regular in coloration. The disease wasfound to have no apparent sex predilection <strong>and</strong> is mostfrequent in <strong>the</strong> 6th <strong>and</strong> 7th decades [319].The floor <strong>of</strong> <strong>the</strong> mouth, <strong>the</strong> ventral <strong>and</strong> lateral tongue,<strong>the</strong> retromolar region <strong>and</strong> <strong>the</strong> s<strong>of</strong>t palate are <strong>the</strong> mostfrequently involved sites [47, 263]. OEs that are intermixedwith white areas are called erythroleukoplakia orspeckled mucosa <strong>and</strong> are believed to behave similarly topure OE. The red appearance <strong>of</strong> OE may be related toan increase in subepi<strong>the</strong>lial blood vessels, a lack <strong>of</strong> surfacekeratin <strong>and</strong> thinness <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium. Prior to aclinical diagnosis <strong>of</strong> OE numerous entities should be excluded,such as: median rhomboid glossitis, all kinds <strong>of</strong>injuries, infectious <strong>and</strong> allergic lesions, haemorrhages,vessel tumours, Wegener’s granulomatosis, etc. [47]. AlthoughOE is a rare lesion, it is much more likely to showdysplasia or carcinoma. Shafer <strong>and</strong> Waldron reviewed<strong>the</strong>ir biopsy experiences with 65 cases <strong>of</strong> OE: 51% <strong>of</strong> casesshowed invasive SCC, 40% were carcinomas in situor severe dysplasia, <strong>and</strong> <strong>the</strong> remaining 9% showed mildto moderate dysplasia [319]. In all red lesions <strong>of</strong> <strong>the</strong> oralmucosa that do not regress within 2 weeks <strong>of</strong> <strong>the</strong> removal<strong>of</strong> possible aetiological factors, biopsy is, <strong>the</strong>refore,m<strong>and</strong>atory.1.2.4.2 Laryngeal<strong>and</strong> HypopharyngealLeukoplakia <strong>and</strong>Chronic LaryngitisSquamous intraepi<strong>the</strong>lial lesions appear mainly along<strong>the</strong> true vocal cords, <strong>and</strong> rarely in o<strong>the</strong>r parts <strong>of</strong> <strong>the</strong> larynx,such as <strong>the</strong> epiglottis. Two-thirds <strong>of</strong> vocal cord lesionsare bilateral [48, 178, 181]. They can extend over<strong>the</strong> free edge <strong>of</strong> <strong>the</strong> vocal cord to its subglottic surface.An origin in, or extension along <strong>the</strong> upper surface <strong>of</strong> <strong>the</strong>vocal cord is less common [181, 194]. The commissuresare rarely involved [48]. Hypopharyngeal lesions arerarely found <strong>and</strong> are poorly defined [364].Laryngeal SILs do not have a single distinctive orcharacteristic clinical appearance <strong>and</strong> are variously describedas leukoplakia, chronic hyperplastic laryngitisor rarely erythroplakia. A circumscribed thickening <strong>of</strong><strong>the</strong> mucosa covered by whitish patches (Fig. 1.7), or anirregularly growing, well-defined warty plaque may beseen. A speckled appearance <strong>of</strong> lesions can also be present,caused by unequal thickness <strong>of</strong> <strong>the</strong> keratin layer.However, <strong>the</strong> lesions are commonly more diffuse,with a thickened appearance, <strong>and</strong> occupy a large part <strong>of</strong>one or both vocal cords (Fig. 1.8). A few leukoplakic lesionsare ulcerated (6.5%) or combined with erythroplakia(15%) [48]. Leukoplakic lesions, in contrast to erythroplakicones, tend to be well demarcated.The macroscopic features <strong>of</strong> hypopharyngeal <strong>and</strong>laryngeal SILs are not as well defined as <strong>the</strong>ir counterpartsin <strong>the</strong> oral cavity <strong>and</strong> <strong>the</strong>ir relative importance isnot generally accepted. Most patients with SILs presentwith a history <strong>of</strong> a few months or more <strong>of</strong> symptoms;an average duration <strong>of</strong> 7 months has been reported[48]. Symptoms depend on <strong>the</strong> location <strong>and</strong> sever-


8 N. Gale · N. Zidar1Fig. 1.7. Leukoplakia <strong>of</strong> <strong>the</strong> left vocal cord. The microscopic diagnosiswas squamous cell hyperplasiaTraditional light microscopic examination, in spite <strong>of</strong>certain subjectivity in interpretation, remains <strong>the</strong> mostreliable method for determining an accurate diagnosis<strong>of</strong> a SIL. The clinical validity <strong>of</strong> any histological gradingsystem depends on <strong>the</strong> degree <strong>of</strong> accord with <strong>the</strong> biologicalbehaviour <strong>of</strong> <strong>the</strong> lesions. Worldwide, <strong>the</strong>re areno generally accepted criteria for a histological gradingsystem in <strong>the</strong> head <strong>and</strong> neck region with regard to severity<strong>of</strong> SILs <strong>and</strong> propensity to malignant transformation.It is, <strong>the</strong>refore, not surprising to find in <strong>the</strong> literaturemore than 20 classifications <strong>of</strong> laryngeal SILs [39,125, 150, 180, 181, 242]. The majority <strong>of</strong> <strong>the</strong> classificationshave followed similar criteria to those in commonuse for epi<strong>the</strong>lial lesions <strong>of</strong> <strong>the</strong> uterine cervix, such as<strong>the</strong> dysplasia or cervical intraepi<strong>the</strong>lial neoplasia systems.The World Health Organisation (WHO) has recentlyreadopted <strong>the</strong> dysplasia system for classifying SILs <strong>of</strong><strong>the</strong> oral cavity <strong>and</strong> larynx [381]. However, due to differentst<strong>and</strong>points concerning this important problem <strong>of</strong>oral <strong>and</strong> laryngeal carcinogenesis, <strong>the</strong> dysplasia systemwas reviewed simultaneously with two additional classifications:<strong>the</strong> squamous intraepi<strong>the</strong>lial neoplasia system<strong>and</strong> <strong>the</strong> Ljubljana classification [381]. Here, <strong>the</strong> WHOdysplasia system <strong>and</strong> <strong>the</strong> Ljubljana classification will bereviewed.Fig. 1.8. Chronic laryngitis. Both vocal cords are irregularlythickened <strong>and</strong> covered by whitish plaques. The microscopic diagnosiswas atypical hyperplasiaity <strong>of</strong> <strong>the</strong> disease. Patients may complain <strong>of</strong> fluctuatinghoarseness, throat irritation, sore throat, <strong>and</strong>/or achronic cough.1.2.5 Histological Classifications1.2.5.1 WHO Dysplasia SystemPrecursor lesions are designated as altered epi<strong>the</strong>liumwith an increased likelihood <strong>of</strong> progression to SCC. Thealtered epi<strong>the</strong>lium shows a variety <strong>of</strong> architectural <strong>and</strong>cytological changes that have been grouped under <strong>the</strong>term dysplasia. The following architectural changes arerequired to diagnose epi<strong>the</strong>lial dysplasia: irregular epi<strong>the</strong>lialstratification, loss <strong>of</strong> polarity <strong>of</strong> basal cells, dropshapedrete ridges, increased number <strong>of</strong> mitoses, superficialmitoses, dyskeratosis <strong>and</strong> keratin pearls withinrete pegs. The cytological abnormalities <strong>of</strong> dysplasia are:anisonucleosis, nuclear pleomorphism, anisocytosis,cellular pleomorphism, increased nuclear cytoplasmicratio, atypical mitotic figures, increased number <strong>and</strong>size <strong>of</strong> nucleoli, <strong>and</strong> hyperchromatism.The dysplasia system includes <strong>the</strong> following categories:A. Hyperplasia with increased number <strong>of</strong> cells. Thismay be in <strong>the</strong> spinous layer (acanthosis) or in <strong>the</strong>basal <strong>and</strong> parabasal cell layer (basal cell hyperplasia).The architecture <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium is preserved, <strong>and</strong><strong>the</strong>re is no cellular atypia.B. Dysplasia with three grades:1. Mild dysplasia: architectural disturbance is limitedto <strong>the</strong> lower third <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium, accompanied bycytological atypia.2. Moderate dysplasia: architectural disturbance extendsinto <strong>the</strong> middle third <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium, accompaniedby an upgraded degree <strong>of</strong> cytological atypia.


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 93. Severe dysplasia: architectural disturbance is greaterthan two-thirds <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium with associatedcytological atypia or architectural disturbance in <strong>the</strong>middle third <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium with sufficient cytologicalatypia to upgrade from moderate to severedysplasia.C. Carcinoma in situ: full or almost full thickness <strong>of</strong> <strong>the</strong>epi<strong>the</strong>lium shows architectural disturbance, accompaniedby pronounced cytological atypia. Atypicalmitotic figures <strong>and</strong> abnormal superficial mitoses arepresent [381].1.2.5.2 The Ljubljana ClassificationThe Ljubljana grading system does not follow <strong>the</strong> criteriaused for cervical SILs, but was devised to cater for <strong>the</strong>special clinical <strong>and</strong> histological problems related to laryngealconditions. Briefly, <strong>the</strong> different aetiology <strong>of</strong> SILs in<strong>the</strong> upper aerodigestive tract in comparison with cervicallesions probably triggers a different pathway <strong>of</strong> geneticevents from those established in cervical lesions. Additionally,different anatomic specificities, various clinicalapproaches to obtaining adequate biopsy specimens, aswell as different treatment modalities for high-risk lesions<strong>of</strong> cervical <strong>and</strong> upper aerodigestive tract SILs, were<strong>the</strong> basis for establishing <strong>the</strong> Ljubljana classification morethan three decades ago <strong>and</strong> this was fur<strong>the</strong>r formulated in1997 by <strong>the</strong> working group on SILs <strong>of</strong> <strong>the</strong> European Society<strong>of</strong> <strong>Pathology</strong> [125, 150, 178, 181, 182, 183, 242].The main feature <strong>of</strong> <strong>the</strong> Ljubljana classification is thatit separates <strong>the</strong> group <strong>of</strong> lesions with a minimal risk <strong>of</strong>progression to invasive carcinoma, including squamous<strong>and</strong> basal-parabasal cell hyperplasia on <strong>the</strong> one h<strong>and</strong>,<strong>and</strong> <strong>the</strong> potentially malignant group, i.e. those lesionsmore likely to progress to invasive carcinoma (atypicalhyperplasia or risky epi<strong>the</strong>lium), on <strong>the</strong> o<strong>the</strong>r. Carcinomain situ is considered a separate entity within <strong>the</strong>spectrum <strong>of</strong> SILs.The general principles <strong>of</strong> <strong>the</strong> classification presented,which hold for all <strong>of</strong> its grades, are <strong>the</strong> following: <strong>the</strong>epi<strong>the</strong>lium is generally thickened, although in a minority<strong>of</strong> cases <strong>the</strong> epi<strong>the</strong>lium may show areas <strong>of</strong> diminishedthickness, but even <strong>the</strong>se cases show basal-parabasal cellhyperplasia; <strong>the</strong> basement membrane is generally preservedat all grades, with no definite evidence <strong>of</strong> evenminimal invasion. The presence <strong>of</strong> a surface keratin layer,which is <strong>of</strong>ten present in all grades <strong>of</strong> SIL, is <strong>of</strong> no importancein this classification.Group <strong>of</strong> Reactive Lesions with a Minimal Risk<strong>of</strong> Progression to Invasive CarcinomaSquamous (simple) hyperplasia is a benign hyperplasticprocess with retention <strong>of</strong> <strong>the</strong> normal architectural <strong>and</strong>cytological pattern <strong>of</strong> <strong>the</strong> squamous epi<strong>the</strong>lium.Fig. 1.9. Squamous cell hyperplasia. Thickened epi<strong>the</strong>lium showsincreased prickle cell layer, <strong>the</strong> basal layer remains unchangedThe epi<strong>the</strong>lium is thickened as a result <strong>of</strong> an increasedprickle cell layer. The cells <strong>of</strong> <strong>the</strong> basal <strong>and</strong> parabasalregion, which comprise one to three layers, remainunchanged. There is no cellular atypia; infrequent, regularmitoses are seen in <strong>the</strong> basal layer (Fig. 1.9).Basal <strong>and</strong> parabasal cell hyperplasia (abnormal hyperplasia)can be defined as a benign augmentation <strong>of</strong>basal <strong>and</strong> parabasal cells in <strong>the</strong> lower part <strong>of</strong> <strong>the</strong> epi<strong>the</strong>liallayer while <strong>the</strong> upper part, containing regular pricklecells, remains unchanged.Stratification <strong>of</strong> <strong>the</strong> laryngeal squamous epi<strong>the</strong>lium,characterised by its layered construction, is seen asa smooth transition from an epi<strong>the</strong>lial layer composed<strong>of</strong> basal cells that are aligned perpendicular to <strong>the</strong> basementmembrane to <strong>the</strong> more superficial part in which<strong>the</strong> prickle cells are orientated horizontal to <strong>the</strong> basementmembrane. Thickened epi<strong>the</strong>lium consists <strong>of</strong> anincreased number <strong>of</strong> basal <strong>and</strong> parabasal cells occupyingup to one-half or occasionally slightly more <strong>of</strong> <strong>the</strong> entireepi<strong>the</strong>lium. These cells do not show significant nuclearchanges <strong>and</strong> are aligned perpendicularly with preservation<strong>of</strong> normal polarity <strong>and</strong> organisation. Basal <strong>and</strong>parabasal cells contain moderately enlarged nuclei <strong>and</strong>uniformly distributed chromatin, slightly more cytoplasmthan those <strong>of</strong> <strong>the</strong> basal layer <strong>and</strong>, in addition, fewor no intercellular prickles or bridges. Rare, regular mitosesmay be seen, always located in or near <strong>the</strong> basal layer.Less than 5% <strong>of</strong> epi<strong>the</strong>lial cells show characteristics <strong>of</strong>


10 N. Gale · N. Zidar1Fig. 1.10. Basal <strong>and</strong> parabasal cell hyperplasia. Augmented cells<strong>of</strong> <strong>the</strong> basal <strong>and</strong> parabasal cell layer extend up to <strong>the</strong> midportion<strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium. Occasional mitoses are seen in <strong>the</strong> lower part <strong>of</strong><strong>the</strong> epi<strong>the</strong>liumadyskeratosis, a premature <strong>and</strong> abnormal keratinisation<strong>of</strong> individual cells or groups <strong>of</strong> cells that have no prickles<strong>and</strong> strongly eosinophilic cytoplasm (Fig. 1.10).Group <strong>of</strong> Potentially Malignant LesionsAtypical hyperplasia (risky epi<strong>the</strong>lium), considered tobe a potentially malignant lesion, i.e. a lesion with adefinitely increased risk <strong>of</strong> progressing to invasive carcinoma,is characterised by preservation <strong>of</strong> stratificationin <strong>the</strong> epi<strong>the</strong>lium, by alterations <strong>of</strong> epi<strong>the</strong>lial cells withmild to moderate degrees <strong>of</strong> cytological atypia occupying<strong>the</strong> lower half or more <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lial thickness,<strong>and</strong> by increased mitotic activity.Stratification is still preserved in <strong>the</strong> epi<strong>the</strong>lium.There is an increased number <strong>of</strong> epi<strong>the</strong>lial cells that arefrequently orientated perpendicular to <strong>the</strong> basementmembrane. The nuclei <strong>of</strong> many <strong>of</strong> <strong>the</strong>m show mild tomoderate changes <strong>of</strong> atypia, such as: enlargement, irregularcontours, <strong>and</strong> marked variations in staining intensitywith frequent hyperchromaticity; nucleoli are increasedin number <strong>and</strong> size, showing enhanced stainingcharacteristics. The nuclear/cytoplasmic ratio is generallyincreased. This type <strong>of</strong> epi<strong>the</strong>lial cell may occupy <strong>the</strong>lower half, or more <strong>of</strong> <strong>the</strong> entire epi<strong>the</strong>lial thickness. Mitosesare moderately increased. They are usually foundin <strong>the</strong> lower two-thirds <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium, although <strong>the</strong>ymay occasionally appear at a higher level. Mitoses arerarely, if ever, abnormal. Dyskeratotic cells are frequentwithin <strong>the</strong> entire epi<strong>the</strong>lium. Apoptotic cells may bepresent; <strong>the</strong>y are smaller in size <strong>and</strong> with hyaline eosinophiliccytoplasm <strong>and</strong> conspicuous nuclear chromatinbFig. 1.11. a Atypical hyperplasia. Augmented epi<strong>the</strong>lial cellsshowing mild to moderate grades <strong>of</strong> atypia, preserved stratification<strong>and</strong> some regular mitoses. b Augmented epi<strong>the</strong>lial cells withincreased nuclear/cytoplasmic ratio <strong>and</strong> some regular mitoses.The cells are aligned perpendicularly to <strong>the</strong> basement membranecondensation or nuclei crumbled into smaller fragments(Fig. 1.11). Two subdivisions <strong>of</strong> atypical hyperplasia arerecognised: (a) The more frequent “basal cell type” withno intercellular prickles <strong>and</strong> no cytoplasmic eosinophilia,<strong>and</strong> <strong>the</strong> cells aligned perpendicularly or at an acuteangle to <strong>the</strong> basement membrane, <strong>and</strong> (b) The less frequent“spinous cell type” (analogous to so-called “keratinisingdysplasia” by Crissman <strong>and</strong> Zarbo [79]) withintercellular prickles <strong>and</strong> increased cytoplasmic eosino-


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 11five per high power field. Abnormal mitoses are frequentlyseen. Hyaline bodies <strong>and</strong> dyskeratotic cellsare present, <strong>of</strong>ten in high numbers (Fig. 1.12).In CIS, as in atypical hyperplasia, <strong>the</strong> lesion may fallwithin one or <strong>the</strong> o<strong>the</strong>r <strong>of</strong> <strong>the</strong> two subdivisions <strong>of</strong> atypicalhyperplasia:a) Basal cell type with no intercellular prickles <strong>and</strong> nocytoplasmic eosinophilia;b) Spinous cell type with intercellular prickles <strong>and</strong> increasedcytoplasmic eosinophilia [125, 150, 242].Fig. 1.12. Carcinoma in situ. The lesion shows loss <strong>of</strong> stratification,malignant cells with increased mitotic activity replace <strong>the</strong>entire epi<strong>the</strong>lial thicknessphilia. The cells may be aligned horizontal to <strong>the</strong> basementmembrane.Group <strong>of</strong> Actually Malignant LesionsThe term squamous cell carcinoma in situ (CIS) is reservedfor lesions showing <strong>the</strong> features <strong>of</strong> carcinomawithout invasion. Three distinct morphologic characteristicsare usually present:a) Loss <strong>of</strong> stratification or maturation <strong>of</strong> <strong>the</strong> epi<strong>the</strong>liumas a whole; however, <strong>the</strong> surface <strong>of</strong> <strong>the</strong> epi<strong>the</strong>liummay be covered by one or at most a few layers <strong>of</strong> compressed,horizontally stratified, <strong>and</strong> sometimes keratinisedcells.b) Epi<strong>the</strong>lial cells may show all <strong>the</strong> cytologic characteristics<strong>of</strong> invasive squamous cell carcinoma.c) Mitotic figures are usually markedly increasedthroughout <strong>the</strong> whole epi<strong>the</strong>lium, <strong>of</strong>ten more thanIn <strong>the</strong> differential diagnosis <strong>of</strong> SILs, regenerative changes<strong>of</strong> <strong>the</strong> squamous epi<strong>the</strong>lium after trauma, inflammationor irradiation <strong>the</strong>rapy may simulate various cytoarchitecturaldisturbances resembling different grades<strong>of</strong> SILs.Clinical data are always <strong>of</strong> considerable help in distinguishingdifferent grades <strong>of</strong> SILs from regenerativeprocesses in which epi<strong>the</strong>lial abnormalities are generallyless pronounced than in atypical hyperplasia or CIS,<strong>and</strong> atypical mitoses are almost never present.1.2.5.3 Comparison Between<strong>the</strong> Ljubljana Classification<strong>and</strong> WHO 2005 ClassificationComparing <strong>the</strong> two classifications, one should be awarethat <strong>the</strong>re is no simple relationship <strong>and</strong> overlapping between<strong>the</strong> WHO 2005 <strong>and</strong> <strong>the</strong> Ljubljana classifications(Table 1.1).The group <strong>of</strong> <strong>the</strong> so-called benign lesions, includingsquamous <strong>and</strong> basal-parabasal cell (abnormal) hyperplasiais comparable in both classifications. Disagreementstarts with <strong>the</strong> presumption <strong>of</strong> <strong>the</strong> WHO 2005 classification[381] that each grade <strong>of</strong> <strong>the</strong> whole series <strong>of</strong> dysplasiais considered to be a precursor or potentially malignantlesion. Histologically, however, <strong>the</strong>re are some similaritiesbetween <strong>the</strong> basal <strong>and</strong> parabasal cell hyperplasia<strong>of</strong> <strong>the</strong> Ljubljana classification <strong>and</strong> <strong>the</strong> mild dysplasia<strong>of</strong> <strong>the</strong> WHO 2005 [381]. Mild dysplasia, in contrast tobasal <strong>and</strong> parabasal cell hyperplasia, was classified as<strong>the</strong> initial grade within a potentially malignant group,Table 1.1. Comparison between two classifications <strong>of</strong> squamous intraepi<strong>the</strong>lial lesions: WHO 2005 <strong>and</strong> Ljubljana classification [381]WHO dysplasia systemHyperplasia*Mild dysplasiaModerate dysplasiaSevere dysplasiaCarcinoma in situLjubljana classification <strong>of</strong> squamous intraepi<strong>the</strong>lial lesionsSquamous cell hyperplasiaHyperplasia <strong>of</strong> basal <strong>and</strong> parabasal cell layersAtypical hyperplasia – risky epi<strong>the</strong>liumCarcinoma in situ*Hyperplasia may be in <strong>the</strong> spinous <strong>and</strong>/or in <strong>the</strong> basal/parabasal cell layers


12 N. Gale · N. Zidar1whereas in <strong>the</strong> Ljubljana classification, basal-parabasalhyperplasia is considered a benign lesion with a minimumrisk <strong>of</strong> malignant transformation. Atypical hyperplasia<strong>of</strong> <strong>the</strong> Ljubljana classification is similar to moderatedysplasia, but also partially includes severe dysplasia.The analogy is, thus, only approximate [150]. Carcinomain situ is equal to <strong>the</strong> carcinoma in situ <strong>of</strong> <strong>the</strong> WHO2005 classification. However, some cases <strong>of</strong> severe dysplasiawould fall into <strong>the</strong> category <strong>of</strong> carcinoma in situ<strong>of</strong> <strong>the</strong> Ljubljana classification, <strong>and</strong> <strong>the</strong> analogy is againonly approximate [150].The Ljubljana classification was devised to satisfy<strong>the</strong> specific clinical <strong>and</strong> histological requirements <strong>of</strong><strong>the</strong> diagnosis <strong>of</strong> SILs in <strong>the</strong> regions <strong>of</strong> <strong>the</strong> upper aerodigestivetract where common aetiological, clinical <strong>and</strong>morphological aspects are found. Recently, <strong>the</strong> Ljubljanaclassification has also been successfully applied tooral SILs, which share <strong>the</strong> same aetiology, <strong>and</strong> similarclinical <strong>and</strong> histological specificities with laryngeal lesions[225].Over <strong>the</strong> many years in practical use, it has beenfound to be more precise for daily diagnostic work thano<strong>the</strong>r grading systems <strong>and</strong> provides data that have beenshown to be closely correlated with <strong>the</strong> biological behaviour<strong>of</strong> <strong>the</strong> lesions [125].1.2.6 Biomarkers Related to MalignantPotential <strong>of</strong> SILs Recognisedby Auxiliary <strong>and</strong> AdvancedMolecular MethodsA genetic progression model with specific genetic alterationsfor different stages <strong>of</strong> laryngeal SILs has increased<strong>the</strong> possibilities <strong>of</strong> recognising potential biomarkers incorrelation with histopathologic changes that mightsignal a stage <strong>of</strong> carcinogenesis from initiation to invasivegrowth [60]. This model has revealed that bothoncogenes <strong>and</strong> tumour suppressor genes are involvedin tumour progression with a distinct order <strong>of</strong> progressionstarting with loss <strong>of</strong> heterozygosity (LOH) at 9p21<strong>and</strong> 3p21 as <strong>the</strong> earliest detectable events, followed by17p13 loss. Additional genetic alterations, which tend tooccur in severe dysplasia (atypical hyperplasia), or evenin SCC, are cyclin D1 amplification, pTEN inactivation,<strong>and</strong> LOH at 11q13, 13q21, 14q32, 6p, 8q, 4q27, <strong>and</strong> 10q23 [60, 117]. For some chromosomal areas involved <strong>the</strong>target genes have been recognised, such as tumour suppressorgenes p16 at 9p21, p53 at 17p13, <strong>and</strong> cyclin D1oncogene at 11q13 [60, 117, 381].A similar genetic basis, associated with histopathologicalstages, has been designed for oral carcinogenesis,based on LOH, gene mutations <strong>and</strong> telomerase reactivation[231]. Recent approaches to identifying geneticchanges as predictors <strong>of</strong> malignancy risk for low gradeoral dysplasia show that LOH at 3p <strong>and</strong> 9p could serveas an initial screening marker for <strong>the</strong> cancer risk <strong>of</strong> earlylesions [306]. Additionally, telomerase reactivation hasbeen shown to be an early event <strong>of</strong> laryngeal <strong>and</strong> oralcarcinogenesis, already detectable at <strong>the</strong> stage <strong>of</strong> atypicalhyperplasia in 75% <strong>and</strong> 43% respectively. However, forprogression towards invasive SCC o<strong>the</strong>r genetic eventsseem to be necessary [225, 226].Special attention has been recently devoted to moleculargenetic studies <strong>of</strong> potentially malignant lesions inan attempt to establish <strong>the</strong>ir risk <strong>of</strong> progression more reliablythan static conventional histological diagnosis enables.In terms <strong>of</strong> prognostic value, genetic events suchas LOH <strong>of</strong> 3p, 9p21 <strong>and</strong> 17q 13 <strong>and</strong> DNA aneuploidy areconsidered a substantial risk <strong>of</strong> malignant transformation[344, 381].Predictive factors <strong>of</strong> different grades <strong>of</strong> SILs inhead <strong>and</strong> neck carcinogenesis have also been widelystudied at <strong>the</strong> level <strong>of</strong> abnormal protein expression<strong>of</strong> <strong>the</strong> oncogenes <strong>and</strong> tumour suppressor genesinvolved. Overexpression <strong>of</strong> p16, p21waf1, p27, p53,epidermal growth factor receptor (EGFR), <strong>and</strong> cyclinD1 proteins have been examined in an attempt to increasediagnostic sensitivity <strong>and</strong> predictive values <strong>of</strong>SILs [12, 64, 102, 127, 156, 162, 169, 199, 251, 255, 282,363, 369].Additionally, various proliferation <strong>and</strong> differentiationmarkers, including keratins <strong>and</strong> carbohydrate antigens,are widely used as predictive factors for determining<strong>the</strong> biological behaviour <strong>of</strong> oral <strong>and</strong> laryngealSILs [297]. The detection <strong>of</strong> proliferative activity, suchas <strong>the</strong> counting <strong>of</strong> nucleolar organiser regions (Ag-NORs) <strong>and</strong> immunohistochemical labelling for proliferatingcell nuclear antigen (PCNA) <strong>and</strong> Ki-67 antigenare useful adjuncts to light microscopy <strong>and</strong> may providepredictive information on <strong>the</strong> clinical outcome <strong>of</strong>SILs in <strong>the</strong> larynx <strong>and</strong> oral cavity [73, 129, 181, 251, 279,357, 394].The expression <strong>of</strong> lectins <strong>and</strong> cytokeratins, particularlythose <strong>of</strong> low molecular weight, has been shown tobe a good marker <strong>of</strong> epi<strong>the</strong>lial maturation in normal <strong>and</strong>pathologic conditions, <strong>and</strong> may thus facilitate a moreprecise evaluation <strong>of</strong> SIL [152, 182, 229, 365].1.2.7 Treatment <strong>and</strong> Prognosis1.2.7.1 Oral Cavity <strong>and</strong> OropharynxSurgical excision, performed ei<strong>the</strong>r classically witha cold knife or a CO 2 laser, is <strong>the</strong> treatment <strong>of</strong> choicefor oral SILs. However, in highly suspicious lesions asin OE on <strong>the</strong> floor <strong>of</strong> <strong>the</strong> mouth, an incisional biopsy isalways <strong>the</strong> preferred method for establishing a microscopicdiagnosis. Surgical treatment is only <strong>the</strong> beginning<strong>of</strong> <strong>the</strong>rapy for such lesions; <strong>the</strong> long-term follow-


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 13up <strong>and</strong> avoidance <strong>of</strong> exposure to known risk factors isimportant due to <strong>the</strong> risk <strong>of</strong> malignant transformation[47, 263, 334]. Recurrences <strong>of</strong> high-risk SILs are not infrequentevents, being reported in 18% <strong>of</strong> lesions thathad been excised with free surgical margins [366]. If <strong>the</strong>size or o<strong>the</strong>r clinical obstacles make surgical treatment<strong>of</strong> oral SILs difficult, various antioxidants, such as betacarotene<strong>and</strong> <strong>the</strong> retinoids, are most commonly used forchemoprevention [191].The occurrence <strong>of</strong> <strong>the</strong> higher grades (moderate <strong>and</strong>severe dysplasia, atypical hyperplasia) <strong>of</strong> oral SILs isconsidered <strong>the</strong> most important risk <strong>of</strong> SCC development.The reported frequency <strong>of</strong> malignant transformation<strong>of</strong> OL ranges from 3.1% [373] to 17.5% [323]. Severallocations <strong>of</strong> OL, toge<strong>the</strong>r with histological abnormalities,are linked with higher malignant transformation.The floor <strong>of</strong> <strong>the</strong> mouth is, thus, <strong>the</strong> highest risk site, followedby <strong>the</strong> tongue <strong>and</strong> lip [319].The clinical appearance <strong>of</strong> non-homogenous or speckledOL may correlate with <strong>the</strong> likelihood that <strong>the</strong> lesionwill show epi<strong>the</strong>lial changes or malignant transformation.In a study by Silverman <strong>and</strong> Gorsky <strong>the</strong> overall malignanttransformation <strong>of</strong> OL was 17.5%, for <strong>the</strong> homogenousform only 6.6%, <strong>and</strong> for speckled OL 23.4%. Aspecial subtype <strong>of</strong> OL, PVL, was found to develop SCCin 70.3% <strong>of</strong> patients [322]. Compared with OL, OE hassignificantly worse biological behaviour, with 51% proceedingto malignant transformation [319].1.2.7.2 LarynxThe main task <strong>of</strong> <strong>the</strong> pathologist dealing with laryngealSILs is to separate non-risky or a minimally riskyfrom risky changes. Patients with benign hyperplasticlesions (simple <strong>and</strong> basal-parabasal hyperplasia) donot require such a close follow-up after excisional biopsiesas those with atypical hyperplasia <strong>and</strong> CIS, althoughelimination <strong>of</strong> known detrimental influencesis advised [125, 150]. Diagnosis <strong>of</strong> atypical hyperplasiain laryngeal lesions requires close follow-up <strong>and</strong> <strong>of</strong>tenrepeated histological assessment to detect any possiblepersistence or progression <strong>of</strong> <strong>the</strong> disease [125, 150, 178,181]. Patients with CIS may require more extensive surgicaltreatment or radio<strong>the</strong>rapy, although this is controversial[79, 181, 254, 299, 336].The histopathologic degree <strong>of</strong> severity <strong>of</strong> laryngealSILs is still used as <strong>the</strong> most reliable predictive factor[39, 125, 150, 178, 181, 239]. The frequency <strong>of</strong> subsequentmalignant alteration markedly increases fromsquamous (simple) <strong>and</strong> basal-parabasal (abnormal) hyperplasia(0.9%), compared with atypical hyperplasia(11 %) [150]. Barnes’s review <strong>of</strong> <strong>the</strong> literature shows that<strong>the</strong> risk <strong>of</strong> SCCs developing in mild, moderate <strong>and</strong> severelaryngeal dysplasia ranges from 5.5% to 22.5% <strong>and</strong>28.4% respectively [20].1.3 Invasive Squamous Cell Carcinoma1.3.1 Microinvasive SquamousCell CarcinomaICD-O:8076/3Microinvasive squamous cell carcinoma (SCC) is a SCCwith invasion beyond <strong>the</strong> epi<strong>the</strong>lial basement membrane,extending into <strong>the</strong> superficial stroma. There is little consensusamong pathologists on <strong>the</strong> maximum depth <strong>of</strong>invasion in microinvasive SCCs, but it generally rangesfrom 0.5 mm [20] to 2 mm [77]. The depth <strong>of</strong> invasionmust be measured from <strong>the</strong> basement membrane <strong>of</strong> <strong>the</strong>adjacent (non-neoplastic) surface epi<strong>the</strong>lium, because <strong>of</strong><strong>the</strong> great variations in epi<strong>the</strong>lial thickness.Microinvasive SCC is a biologically malignant lesioncapable <strong>of</strong> gaining access to lymphatic <strong>and</strong> blood vessels,which may result in metastases. However, metastasesare rare in microinvasive SCCs <strong>and</strong> <strong>the</strong> prognosisis excellent. Studies on SCCs <strong>of</strong> <strong>the</strong> floor <strong>of</strong> <strong>the</strong> mouthhave shown that <strong>the</strong>re is little or even no metastatic potentialfor SCCs penetrating less than 2 mm beyond <strong>the</strong>basement membrane, <strong>and</strong> a substantially higher risk <strong>of</strong>metastases in more deeply invasive SCCs at this site [74,77, 246]. The prognosis is also excellent in microinvasiveSCCs <strong>of</strong> <strong>the</strong> laryngeal glottis because <strong>of</strong> <strong>the</strong> poor lymphatic<strong>and</strong> vascular network in this location. Some authorshave <strong>the</strong>refore recommended more conservativetreatment <strong>of</strong> <strong>the</strong>se lesions, such as endoscopic removal,with a careful follow-up [80, 308, 341].The reliable diagnosis <strong>of</strong> microinvasive SCC can onlybe made with certainty if <strong>the</strong> whole lesion is examined.It should not be made in small, tangentially cut biopsyspecimens.1.3.2 Conventional SquamousCell CarcinomaICD-O:8070/3Squamous cell carcinoma (SCC) is a malignant epi<strong>the</strong>lialtumour with evidence <strong>of</strong> squamous differentiationsuch as intercellular bridges <strong>and</strong> keratin formation. Itoriginates from <strong>the</strong> surface squamous epi<strong>the</strong>lium, orfrom ciliated respiratory epi<strong>the</strong>lium that has undergonesquamous metaplasia [242].Squamous cell carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck is <strong>the</strong>sixth most prevalent cancer worldwide, accounting for5% <strong>of</strong> all new cancers, with a global annual incidence<strong>of</strong> 500,000 [42]. The vast majority <strong>of</strong> SCCs are <strong>the</strong> conventionaltype <strong>of</strong> SCC, accounting for more than 90%<strong>of</strong> cases. The remaining cases belong to <strong>the</strong> variants <strong>of</strong>SCC, which will be discussed later in this chapter.Squamous cell carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck occursmost frequently in <strong>the</strong> oral cavity <strong>and</strong> lip, in <strong>the</strong>


14 N. Gale · N. Zidar1a b cFig. 1.13. a Well-differentiated squamous cell carcinoma. b Moderatelydifferentiated squamous cell carcinoma. c Poorly differentiatedsquamous cell carcinomaoropharynx, larynx <strong>and</strong> hypopharynx. Less frequently,it arises in <strong>the</strong> nasopharynx, nasal cavities <strong>and</strong> paranasalsinuses. The predilection sites in <strong>the</strong> oral cavityare <strong>the</strong> lateral tongue <strong>and</strong> floor <strong>of</strong> <strong>the</strong> mouth. In<strong>the</strong> oropharynx, <strong>the</strong> most commonly involved sites are<strong>the</strong> base <strong>of</strong> <strong>the</strong> tongue <strong>and</strong> <strong>the</strong> tonsils. In <strong>the</strong> larynx,<strong>the</strong>re are geographic differences in <strong>the</strong> topographicdistribution, <strong>the</strong> glottis being <strong>the</strong> most frequent locationin some countries, <strong>and</strong> <strong>the</strong> supraglottis in o<strong>the</strong>rs[20, 117].1.3.2.1 AetiologySmoking <strong>and</strong> alcohol abuse are <strong>the</strong> greatest risk factorsfor <strong>the</strong> development <strong>of</strong> SCCs <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Muchattention has been paid to <strong>the</strong> possible role <strong>of</strong> viral infection,particularly <strong>the</strong> Epstein-Barr virus (EBV), <strong>and</strong><strong>the</strong> human papillomavirus (HPV), in <strong>the</strong> pathogenesis<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck carcinoma.The EBV is aetiopathogenetically strongly related tonasopharyngeal carcinomas [265], <strong>and</strong> to rare cases <strong>of</strong>lymphoepi<strong>the</strong>lial carcinoma <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s [153,160]. HPV has been aetiologically linked to SCCs <strong>of</strong> <strong>the</strong>tonsil [97, 214]. Apart from tonsillar SCC <strong>and</strong> nasopharyngealSCC, it appears that EBV <strong>and</strong> HPV play little, ifany, role in <strong>the</strong> pathogenesis <strong>of</strong> SCCs in o<strong>the</strong>r locationsin <strong>the</strong> head <strong>and</strong> neck [93, 153, 227, 392].1.3.2.2 Pathologic FeaturesThe macroscopic appearance <strong>of</strong> invasive SCCs is variable,<strong>and</strong> includes flat lesions with a well-defined, raisededge, polypoid exophytic <strong>and</strong> papillary lesions, as wellas endophytic infiltrative lesions. The surface <strong>of</strong> <strong>the</strong> tumouris frequently ulcerated.Microscopically, SCCs are characterised by an invasivegrowth <strong>and</strong> evidence <strong>of</strong> squamous differentiation.Invasive growth is manifested by interruption <strong>of</strong> <strong>the</strong>basement membrane <strong>and</strong> <strong>the</strong> growth <strong>of</strong> isl<strong>and</strong>s, cords,or single (dyscohesive) tumour cells in <strong>the</strong> subepi<strong>the</strong>lialstroma; large tumours may invade deeper structures,i.e. muscle, cartilage <strong>and</strong> bone. Perineural invasion <strong>and</strong>invasion <strong>of</strong> lymphatic <strong>and</strong> blood vessels may be present<strong>and</strong> are reliable pro<strong>of</strong> <strong>of</strong> invasive cancer. Squamousdifferentiation is demonstrated by intercellular bridges<strong>and</strong>/or keratinisation, with keratin pearl formation.Immunohistochemically, SCCs express epi<strong>the</strong>lialmarkers, such as cytokeratins <strong>and</strong> epi<strong>the</strong>lial membraneantigen (EMA). The patterns <strong>of</strong> expression <strong>of</strong> cytokeratinsubtypes are related to <strong>the</strong> degree <strong>of</strong> SCC differentiation<strong>and</strong> to <strong>the</strong> degree <strong>of</strong> keratinisation [229].The pattern <strong>of</strong> cytokeratin expression in low-gradeSCCs is similar to that observed in non-neoplastic squamousepi<strong>the</strong>lium, <strong>and</strong> is characterised by medium <strong>and</strong>high molecular weight cytokeratins, <strong>and</strong> <strong>the</strong> lack <strong>of</strong> expression<strong>of</strong> <strong>the</strong> low molecular weight cytokeratins. HighgradeSCCs tend to lose <strong>the</strong> expression <strong>of</strong> medium <strong>and</strong>high molecular weight cytokeratins <strong>and</strong> express low molecularweight cytokeratins [229].Of <strong>the</strong> various cytokeratin subtypes, cytokeratins 8,18 <strong>and</strong> 19, recognised by <strong>the</strong> antibody CAM5.2, couldbe used as an indicator <strong>of</strong> malignant transformation.In a study by Mall<strong>of</strong>ré et al., 40% <strong>of</strong> SCCs were positivefor CAM5.2, but it was never positive in non-neoplasticsquamous epi<strong>the</strong>lium [229]. In poorly differentiatedSCCs, expression <strong>of</strong> vimentin may appear [367].


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 151.3.2.3 GradingSquamous cell carcinomas are traditionally graded intowell-, moderately, <strong>and</strong> poorly differentiated SCC. Thecriteria for grading are: <strong>the</strong> degree <strong>of</strong> differentiation,nuclear pleomorphism <strong>and</strong> mitotic activity. Well-differentiatedSCCs closely resemble normal squamousepi<strong>the</strong>lium <strong>and</strong> contain varying proportions <strong>of</strong> large,differentiated keratinocyte-like squamous cells, <strong>and</strong>small basal-type cells, which are usually located at <strong>the</strong>periphery <strong>of</strong> <strong>the</strong> tumour isl<strong>and</strong>s. There are intercellularbridges <strong>and</strong> usually full keratinisation; mitoses arescanty (Fig. 1.13a). Moderately differentiated SCCs exhibitmore nuclear pleomorphism <strong>and</strong> more mitoses, includingabnormal mitoses; <strong>the</strong>re is usually less keratinisation(Fig. 1.13b). In poorly differentiated SCCs, basaltypecells predominate, with a high mitotic rate, includingabnormal mitoses, barely discernible intercellularbridges <strong>and</strong> minimal, if any, keratinisation (Fig. 1.13c).Although keratinisation is more likely to be present inwell- or moderately differentiated SCCs, it should not beconsidered an important histological criterion for gradingSCCs.1.3.2.4 Invasive FrontTumour growth at <strong>the</strong> invasive front (tumour–host interface)shows an expansive pattern, an infiltrative pattern,or both. An expansive growth pattern is characterisedby large tumour isl<strong>and</strong>s with well-defined pushingmargins, whereas an infiltrative pattern is characterisedby small scattered irregular cords or single tumour cells,with poorly defined infiltrating margins. It has beendemonstrated that <strong>the</strong> growth pattern at <strong>the</strong> invasivefront has prognostic implications: an infiltrative patternis associated with a more aggressive course <strong>and</strong> poorerprognosis than an expansive pattern [57, 58, 76, 382].1.3.2.5 Stromal ReactionInvasive SCCs are almost always associated with a desmoplasticstromal reaction, which consists <strong>of</strong> proliferation<strong>of</strong> my<strong>of</strong>ibroblasts, excessive deposition <strong>of</strong> extracellularmatrix <strong>and</strong> neovascularisation [63, 94, 221]. Inour experience, desmoplastic stromal reaction is presentonly in invasive SCCs <strong>and</strong> never in SILs, regardless<strong>of</strong> <strong>the</strong>ir grade, <strong>and</strong> may be considered as an additionalmarker <strong>of</strong> invasion [395, 396].Desmoplastic stromal reaction tends to be pronouncedin well- <strong>and</strong> moderately differentiated SCCs<strong>and</strong> weak or absent in poorly differentiated SCCs <strong>and</strong> inlymphoepi<strong>the</strong>lial carcinomas. The intensity <strong>of</strong> desmoplasiais inversely related to <strong>the</strong> density <strong>of</strong> stromal lymphocyticinfiltration. In SCCs with marked desmoplasia,lymphocytic infiltration is focal <strong>and</strong> scarce, whileintense lymphocytic infiltration is found in SCCs withlittle or no desmoplasia.1.3.2.6 Differential DiagnosisThe diagnosis <strong>of</strong> SCC must be confirmed by a biopsy,which must be taken from <strong>the</strong> clinically most suspiciousarea, avoiding <strong>the</strong> central necrotic area. In well-oriented,adequate biopsy samples, <strong>the</strong> diagnosis does usuallynot present a diagnostic problem, as evidence <strong>of</strong> invasivegrowth <strong>and</strong> <strong>of</strong> squamous differentiation is easily found.However, well-differentiated SCCs must be distinguishedfrom verrucous carcinomas <strong>and</strong> papillary SCCs,as well as from benign conditions, such as pseudoepi<strong>the</strong>liomatoushyperplasia. Verrucous carcinomas lack atypia,which are always present in SCCs. Papillary SCCs arecharacterised by papillae formation. which is not <strong>the</strong>prevailing feature in conventional SCCs.Pseudoepi<strong>the</strong>liomatous hyperplasia is a benign conditionassociated with granular cell tumours, mycoticinfection or tuberculosis. It consists <strong>of</strong> deep irregulartongues <strong>and</strong> rete pegs, but <strong>the</strong>re are no abnormal mitosesor atypia, as in SCCs. Identifying <strong>the</strong> associated condition(granular cell tumour or infection) may be helpfulin establishing <strong>the</strong> diagnosis <strong>of</strong> pseudoepi<strong>the</strong>liomatoushyperplasia.Poorly differentiated SCCs must be differentiatedfrom malignant melanomas, malignant lymphomas,neuroendocrine carcinomas, adenocarcinomas, <strong>and</strong> adenosquamouscarcinomas. The correct diagnosis is bestachieved by <strong>the</strong> use <strong>of</strong> appropriate immunohistochemistry<strong>and</strong> special stains for <strong>the</strong> demonstration <strong>of</strong> mucinproduction.Malignant melanomas are distinguished from SCCsby <strong>the</strong> expression <strong>of</strong> S-100, HMB-45 <strong>and</strong> melan-A. Neuroendocrinecarcinomas express neuroendocrine markers(synaptophysin, chromogranin) <strong>and</strong> do not show evidence<strong>of</strong> squamous differentiation, while SCCs do notexpress neuroendocrine markers. Malignant lymphomasare differentiated from SCCs by <strong>the</strong> presence <strong>of</strong> leukocytecommon antigen, <strong>and</strong> markers <strong>of</strong> B- or T-celldifferentiation. Adenocarcinomas <strong>and</strong> adenosquamouscarcinomas can be distinguished from SCCs by <strong>the</strong> presence<strong>of</strong> gl<strong>and</strong>s <strong>and</strong> mucin secretion within <strong>the</strong> tumourcells.1.3.2.7 Treatment <strong>and</strong> PrognosisSquamous cell carcinomas <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck havean overall death risk <strong>of</strong> 40% [328]. The most importantprognostic factor is <strong>the</strong> TNM stage based on <strong>the</strong> size <strong>of</strong><strong>the</strong> primary tumour, <strong>the</strong> presence <strong>of</strong> regional lymphnode metastases, <strong>and</strong> distant metastases [332].


16 N. Gale · N. Zidar1a b cFig. 1.14. Spindle cell carcinoma. a Squamous cell carcinoma inassociation with pleomorphic spindle cells. b Pure spindle cellcomponent: pleomorphic cells with large hyperchromatic nuclei. cPositive staining for cytokeratin in spindle cellsAdditional important prognostic features are: localisation<strong>and</strong> depth <strong>of</strong> <strong>the</strong> tumour [74, 77, 130, 233, 246],presence <strong>of</strong> extracapsular spread in lymph node metastases[85, 109, 155, 335], <strong>and</strong> pattern <strong>of</strong> tumour growth at<strong>the</strong> invasive front [57, 58, 76, 382].The prognostic value <strong>of</strong> some o<strong>the</strong>r parameters, i.e.differentiation <strong>of</strong> <strong>the</strong> tumour [166, 280, 376] <strong>and</strong> DNAploidy [23, 98, 350, 378], is controversial.The treatment <strong>of</strong> choice is complete excision <strong>of</strong> <strong>the</strong>tumour. For small tumours at some locations, such as<strong>the</strong> glottic larynx, <strong>the</strong> primary treatment is radiation. Inlarge tumours, surgery is usually followed by radio<strong>the</strong>rapy.Patients with advanced, unresectable tumours, withor without metastases, are treated by concurrent chemo<strong>the</strong>rapy<strong>and</strong> radio<strong>the</strong>rapy [117].giving rise to both epi<strong>the</strong>lial <strong>and</strong> mesenchymal components[66, 354].1.3.3.1 AetiologySimilar to conventional SCCs, SpCCs have been aetiologicallyrelated to cigarette smoking <strong>and</strong> alcohol consumption[356]. It has been suggested that SpCCs maydevelop after radiation exposure; however, some authorsbelieve that this is not a major aetiologic factor [356].The reported incidence <strong>of</strong> radiation-induced SpCCs <strong>of</strong><strong>the</strong> head <strong>and</strong> neck is between 7.7 <strong>and</strong> 9.1%; <strong>the</strong>y developafter a latent period <strong>of</strong> 1.2 to 16 years after radiation exposure[210, 356].1.3.3 Spindle Cell CarcinomaICD-O:8074/3Spindle cell carcinoma (SpCC) is a biphasic tumourcomposed <strong>of</strong> conventional SCC <strong>and</strong> a malignant spindlecell component. Synonyms for SpCC are sarcomatoidcarcinoma, carcinosarcoma, collision tumour <strong>and</strong> pseudosarcoma.It has been described in various sites <strong>of</strong> <strong>the</strong> body including<strong>the</strong> upper <strong>and</strong> lower respiratory tract, breast,skin, urogenital <strong>and</strong> gastrointestinal tracts, <strong>and</strong> salivarygl<strong>and</strong>s [31]. In <strong>the</strong> head <strong>and</strong> neck, SpCC occurs most frequentlyin <strong>the</strong> larynx [36, 108, 213, 356] <strong>and</strong> oral cavity[13, 96, 304], followed by <strong>the</strong> skin, tonsils, sinonasaltract <strong>and</strong> <strong>the</strong> pharynx [13, 375].The histogenesis <strong>of</strong> this tumour is controversial, but<strong>the</strong>re is mounting evidence that SpCC is a monoclonalneoplasm originating from a non-committed stem cell1.3.3.2 Pathologic FeaturesMacroscopically, SpCCs are usually exophytic polypoidor pedunculated tumours, with frequent surface ulceration.Less <strong>of</strong>ten, SpCCs manifest as sessile, endophyticor ulcero-infiltrative tumours [31, 356].Microscopically, SpCCs consist <strong>of</strong> a SCC component<strong>and</strong> a spindle cell component. The former is representedby in situ carcinoma or by an invasive SCC, <strong>and</strong> is <strong>of</strong>tensmall, requiring multiple sections for demonstration(Fig. 1.14a) [210].The spindle cell component usually forms <strong>the</strong> bulk<strong>of</strong> <strong>the</strong> tumour. Spindle cells are <strong>of</strong>ten pleomorphic, withlarge hyperchromatic nuclei, prominent nucleoli, <strong>and</strong>numerous mitoses (Fig. 1.14b). They are arranged in fasciclesor whorls <strong>and</strong> can assume many histologic patterns,<strong>the</strong> most common being that <strong>of</strong> a malignant fibroushistiocytoma or fibrosarcoma [213, 356]. Foci <strong>of</strong>


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 17osteosarcomatous, chondrosarcomatous, or rhabdosarcomatousdifferentiation may be present, particularlyin patients who had been previously treated by radio<strong>the</strong>rapy[203, 213, 356]. Sometimes, only spindle cellsare present; in such cases, a SpCC can be mistaken fora true sarcoma.However, occasional cases <strong>of</strong> SpCC may be less cellular,closely resembling a reactive fibroblastic proliferation<strong>and</strong> can thus be mistaken for a pseudosarcomatousreaction in a SCC or for radiation-induced stromalatypia [13].Metastases usually contain SCCs alone or both SCC<strong>and</strong> spindle cell components, <strong>and</strong> rarely, only a spindlecell component [205, 232, 340].Electron microscopy has revealed evidence <strong>of</strong> epi<strong>the</strong>lialdifferentiation in spindle cells, such as desmosomes<strong>and</strong> ton<strong>of</strong>ilaments [33, 151, 349, 391].Immunohistochemically, tumour cells in SpCC <strong>of</strong>tenexpress epi<strong>the</strong>lial <strong>and</strong> mesenchymal markers; moreover,keratin <strong>and</strong> vimentin coexpression has been observedon individual tumour cells [241, 391]. Cytokeratin expressioncan be demonstrated in spindle cells in 40–85.7% <strong>of</strong> cases (Fig. 1.14c), depending on <strong>the</strong> number <strong>of</strong>antikeratin antibodies used [96, 241, 329, 349, 360, 391].The most sensitive/reliable epi<strong>the</strong>lial (keratin) markersfor SpCC seem to be keratin (AE1/AE3, K1) K1, K18 <strong>and</strong>epi<strong>the</strong>lial membrane antigen (EMA) [356].Spindle cells always express vimentin <strong>and</strong> <strong>of</strong>ten o<strong>the</strong>rmesenchymal filaments, such as myogenic markers(smooth muscle actin, muscle specific actin, desmin).The presence <strong>of</strong> S-100 protein has been reported in rarecases <strong>of</strong> SpCC [356]. SpCCs do not express glial-fibrillaryacid protein (GFAP), chromogranin or HMB-45[356]. p63 has been recently suggested as an alternativeepi<strong>the</strong>lial marker in SpCCs [213a].1.3.3.3 Differential DiagnosisThe diagnosis <strong>of</strong> a SpCC is based on <strong>the</strong> demonstration<strong>of</strong> an invasive or in situ SCC <strong>and</strong> a malignant spindle cellcomponent. However, when a SCC component cannotbe demonstrated, <strong>the</strong> diagnosis is more difficult, <strong>and</strong> <strong>the</strong>SpCC must be distinguished from a number <strong>of</strong> benign<strong>and</strong> malignant processes, such as spindle cell sarcomas,nodular fasciitis, inflammatory my<strong>of</strong>ibroblastic tumour<strong>and</strong> malignant melanoma.In <strong>the</strong> head <strong>and</strong> neck, true sarcomas (with <strong>the</strong> exclusion<strong>of</strong> chondrosarcomas) <strong>and</strong> benign mesenchymal tumoursare very rare; if present, <strong>the</strong>y are usually locatedin deep structures [356]. It is <strong>the</strong>refore a general viewthat in <strong>the</strong> mucosa <strong>of</strong> <strong>the</strong> upper aerodigestive tract a malignantspindle cell tumour is probably a SpCC <strong>and</strong> nota sarcoma.Negative reaction for S-100 protein <strong>and</strong> HMB45 helpsto distinguish SpCCs from malignant melanomas [96].1.3.3.4 Treatment<strong>and</strong> PrognosisWide surgical excision, alone or with radical neckdissection is <strong>the</strong> most successful treatment for SpCC.Radiation <strong>the</strong>rapy is generally considered less effective.The prognosis is similar to that for conventionalSCCs <strong>and</strong> depends on <strong>the</strong> location <strong>of</strong> <strong>the</strong> tumour <strong>and</strong> <strong>the</strong>stage: glottic SpCCs have a good prognosis, while SpCCsin <strong>the</strong> oral cavity <strong>and</strong> paranasal sinuses behave more aggressively[29, 31]. Prognostic significance has been alsosuggested for <strong>the</strong> gross appearance <strong>of</strong> <strong>the</strong> tumour, i.e.polypoid lesions having a better prognosis than flat ulcerativetumours [375].The reported 5-year survival is between 63 <strong>and</strong>94%; <strong>the</strong> overall lethality <strong>of</strong> <strong>the</strong> tumour is 30–34% [29,356].1.3.4 Verrucous CarcinomaICD-O:8051/3Verrucous carcinoma (VC; Ackerman’s tumour) is avariant <strong>of</strong> well-differentiated SCC that was originallydescribed by Ackerman in 1948 [5]. It is characterisedby an exophytic warty growth that is slowly but locallyinvasive <strong>and</strong> can cause extensive local destruction if leftuntreated. It rarely, if ever, metastasises.The majority <strong>of</strong> VCs (75%) occurs in <strong>the</strong> oral cavity<strong>and</strong> 15% in <strong>the</strong> larynx. In <strong>the</strong> oral cavity, <strong>the</strong> buccal mucosa<strong>and</strong> gingiva are most frequently involved, <strong>and</strong> in<strong>the</strong> larynx, <strong>the</strong> most frequent site <strong>of</strong> occurrence is <strong>the</strong>vocal cords. It rarely occurs in o<strong>the</strong>r locations in <strong>the</strong>head <strong>and</strong> neck, such as <strong>the</strong> nasal cavity, sinonasal tract<strong>and</strong> nasopharynx. It has been also described elsewherein <strong>the</strong> body, i.e. <strong>the</strong> skin, anus, genitalia, urinary bladder<strong>and</strong> oesophagus [339].1.3.4.1 AetiologyVerrucous carcinomas have been aetiologically relatedto <strong>the</strong> use <strong>of</strong> chewing tobacco or snuff. The habitualchewing <strong>of</strong> “pan”, a mixture <strong>of</strong> betel leaf, lime, betelnuts <strong>and</strong> tobacco has been implicated in <strong>the</strong> high incidence<strong>of</strong> VC <strong>of</strong> <strong>the</strong> oral cavity in India [197]. However,tobacco usage is not a reasonable explanation for VC in<strong>the</strong> skin, genitourinary tract <strong>and</strong> o<strong>the</strong>r non-aerodigestivesites [107].A possible aetiologic factor is also human papillomavirus(HPV), as HPV types 16 <strong>and</strong> 18, <strong>and</strong> rarely 6 <strong>and</strong>11, have been found in some, but not all cases <strong>of</strong> VC [52,56, 116, 172, 190].


18 N. Gale · N. Zidar1Fig. 1.15. Verrucous carcinoma. a Projections<strong>and</strong> invaginations lined by thick, welldifferentiatedsquamous epi<strong>the</strong>lium withmarked surface keratinisation, invading <strong>the</strong>stroma with well-defined pushing margins.b Squamous epi<strong>the</strong>lial cells are large <strong>and</strong>lack <strong>the</strong> usual cytologic criteria <strong>of</strong> malignancy.There are numerous dyskeratotic cellsab1.3.4.2 Pathologic FeaturesMacroscopically, VC usually presents as a large, broadbased exophytic tumour with a white keratotic <strong>and</strong>warty surface. On <strong>the</strong> cut surface, it is firm or hard, tanto white, <strong>and</strong> may show keratin-filled surface clefts. It isusually large by <strong>the</strong> time <strong>of</strong> diagnosis, measuring up to10 cm in its greatest dimension.Microscopically, VCs consist <strong>of</strong> thickened clubshapedfiliform projections lined with thick, well-differentiatedsquamous epi<strong>the</strong>lium with marked surface keratinisation(“church-spire” keratosis). The squamousepi<strong>the</strong>lial cells in VCs are large [71] <strong>and</strong> lack <strong>the</strong> usualcytologic criteria <strong>of</strong> malignancy. Mitoses are rare, <strong>and</strong>are only observed in <strong>the</strong> suprabasal layer; <strong>the</strong>re are noabnormal mitoses. VCs invade <strong>the</strong> subjacent stromawith well-defined pushing ra<strong>the</strong>r than infiltrative borders(Fig. 1.15). A lymphoplasmacytic inflammatory responseis common in <strong>the</strong> stroma.Hybrid (mixed) tumours also exist composed <strong>of</strong> VC<strong>and</strong> conventional well-differentiated SCC; <strong>the</strong> reportedincidence for <strong>the</strong> oral cavity <strong>and</strong> <strong>the</strong> larynx is 20 <strong>and</strong>10% [274] respectively. It is important to recognise suchhybrid tumours as foci <strong>of</strong> conventional SCC in an o<strong>the</strong>rwisetypical VC indicate a potential for metastasis. Orvidaset al. reported that a patient with a hybrid carcinoma<strong>of</strong> <strong>the</strong> larynx died <strong>of</strong> <strong>the</strong> disease [274]. Patients with hybridcarcinomas must be treated aggressively as if <strong>the</strong>yhad conventional SCCs [274].Verrucous carcinoma is characterised by a high frequency<strong>of</strong> initial misdiagnosis; Orvidas et al. reporteda series <strong>of</strong> 53 laryngeal VCs; 16 out <strong>of</strong> 31 patients(52%) had received an incorrect diagnosis <strong>of</strong> a benignlesion [274]. This emphasises <strong>the</strong> need for close cooperationbetween <strong>the</strong> pathologist <strong>and</strong> <strong>the</strong> clinician in orderto establish <strong>the</strong> diagnosis <strong>of</strong> VC. An adequate, fullthicknessbiopsy specimen must be taken when a cliniciansuspects a VC [274]; moreover, multiple biopsiesmay be needed to rule out a conventional SCC componentin a VC.1.3.4.3 Differential DiagnosisDifferential diagnosis includes verrucous hyperplasia,well-differentiated SCC, papillary SCC, <strong>and</strong> squamouspapilloma.Invasion below <strong>the</strong> level <strong>of</strong> <strong>the</strong> basal cell layer <strong>of</strong> <strong>the</strong>neighbouring normal squamous epi<strong>the</strong>lium distinguishesVC from verrucous hyperplasia. Whe<strong>the</strong>r this feature,however, adequately discriminates between VC <strong>and</strong> verrucoushyperplasia is debatable, as verrucous hyperplasiacould be an exophytic form <strong>of</strong> VC as well [327].Lack <strong>of</strong> atypia helps to rule out <strong>the</strong> conventional SCC<strong>and</strong> papillary SCC. The VC also lacks <strong>the</strong> well-formed,wide papillary fronds <strong>of</strong> a squamous cell papilloma.An additional feature supporting <strong>the</strong> diagnosis <strong>of</strong>a VC is <strong>the</strong> enlarged spinous cells by morphometricalanalysis [71].1.3.4.4 TreatmentVerrucous carcinoma may be treated by excision (by laseror surgery), <strong>and</strong> by radio<strong>the</strong>rapy. It appears that surgeryis a more effective treatment for VC [236, 274]. Hagenet al. reported a 92.4% cure rate for primary surgeryin patients with laryngeal VC [145]. In contrast, Ferlito<strong>and</strong> Recher reported a 29% cure rate for radio<strong>the</strong>rapy inlaryngeal VC [107]. O<strong>the</strong>r studies have shown a 46–57%rate <strong>of</strong> failure for primary radiation <strong>the</strong>rapy in VCs [224,243, 353].Fur<strong>the</strong>rmore, early reports suggested anaplastictransformation following radio<strong>the</strong>rapy [95, 107, 145,


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 19Fig. 1.16. Papillary squamous cell carcinoma.a Tumour consists <strong>of</strong> papillae with acentral fibrovascular core, covered by neoplasticsquamous epi<strong>the</strong>lium. b The coveringepi<strong>the</strong>lium is composed <strong>of</strong> pleomorphic cellsresembling carcinoma in situMacroscopically, PSCCs present as papillary, friable<strong>and</strong> s<strong>of</strong>t tumours, ranging in size from 2 mm to 4 cm.The main histologic feature <strong>of</strong> PSCCs is <strong>the</strong> papillarygrowth pattern that comprises <strong>the</strong> majority <strong>of</strong> <strong>the</strong> tumour(Fig. 1.16a). Papillae consist <strong>of</strong> a central fibrovascularcore covered by neoplastic squamous epi<strong>the</strong>lium.The covering epi<strong>the</strong>lium may be composed <strong>of</strong> immaturebasaloid cells or may be more pleomorphic, resemblingcarcinoma in situ (Fig. 1.16b). It is usually non-keratinisingor minimally keratinising.Multiple lesions can be found, consisting ei<strong>the</strong>r <strong>of</strong> invasivePSCCs or mucosal papillary hyperplasia. Stromalinvasion is <strong>of</strong>ten difficult to demonstrate in biopsy specimens,<strong>and</strong> sometimes additional biopsies are needed tomake <strong>the</strong> diagnosis <strong>of</strong> an invasive PSCC. A dense lymab278, 287, 309]. However, recent studies do not supportthis notion. It appears that some <strong>of</strong> <strong>the</strong> reported cases <strong>of</strong>transformation <strong>of</strong> VC to SCC after radio<strong>the</strong>rapy were <strong>of</strong>mixed (hybrid) tumours. Moreover, similar transformationcan also occur after surgical treatment <strong>of</strong> VC [237,240, 275, 353]. Radio<strong>the</strong>rapy is now believed to be an appropriatemode <strong>of</strong> treatment for oral VC [177] <strong>and</strong> laryngealVC [275].1.3.4.5 PrognosisIn his original report, Ackerman noted metastasis in <strong>the</strong>regional lymph node in only one <strong>of</strong> <strong>the</strong> 31 patients, <strong>and</strong>no distant spread was observed in his series [5]. Fur<strong>the</strong>rstudies confirmed his observation that pure VCs do notmetastasise [107, 274]; cases <strong>of</strong> VC with metastases werereally a hybrid carcinoma that had not been detected atinitial biopsy.Verrucous carcinomas <strong>the</strong>refore have a good prognosis;<strong>the</strong> overall 5-year survival rate is 77% [196]. It is importantto recognise hybrid tumours, as foci <strong>of</strong> a conventionalSCC in a VC indicate <strong>the</strong> potential for metastasis.Orvidas et al. reported that a patient with a hybridcarcinoma <strong>of</strong> <strong>the</strong> larynx died <strong>of</strong> <strong>the</strong> disease [274]. Patientswith hybrid carcinomas must be treated aggressivelyas if <strong>the</strong>y had a conventional SCC [274].1.3.5 Papillary Squamous Cell CarcinomaICD-O:8052/3Papillary squamous cell carcinoma (PSCC) is an uncommonvariant <strong>of</strong> SCC originally described by Crissman etal. in 1988 [75]. Its main characteristics are a papillarygrowth pattern <strong>and</strong> a good prognosis.In <strong>the</strong> head <strong>and</strong> neck, PSCCs show a predilection for<strong>the</strong> oropharynx, hypopharynx, larynx, <strong>and</strong> <strong>the</strong> sinonasaltract [75, 99, 110, 161, 343, 355]. They also occur ino<strong>the</strong>r parts <strong>of</strong> <strong>the</strong> body, such as <strong>the</strong> skin [15], uterine cervix[292], conjunctiva [215], <strong>and</strong> thymus [209].1.3.5.1 AetiologyIt has been postulated that human papillomavirus(HPV) infection might be an important aetiologic factorin PSCCs, similar to squamous papillomas [343]. However,<strong>the</strong> reported prevalence <strong>of</strong> HPV infection in PSCCsvaries from 0 [75] to 48% [30, 343] <strong>and</strong> does not differsignificantly from <strong>the</strong> reported overall prevalence <strong>of</strong>HPV infection in head <strong>and</strong> neck SCCs [238]. Therefore,<strong>the</strong> significance <strong>of</strong> HPV infection in <strong>the</strong> pathogenesis <strong>of</strong>PSCCs remains unclear.1.3.5.2 Pathologic Features


20 N. Gale · N. Zidar1phoplasmacellular infiltration is usually present in <strong>the</strong>stroma at <strong>the</strong> base <strong>of</strong> <strong>the</strong> carcinoma, but is scarce within<strong>the</strong> papillae. If no stromal invasion is found, <strong>the</strong> lesionis called papillary atypical hyperplasia, PSCC in situ, ornon-invasive PSCC [328].1.3.5.3 Differential DiagnosisDifferential diagnosis includes squamous papilloma,VC, <strong>and</strong> SCC with an exophytic or fungating pattern.Papillomas <strong>and</strong> VCs share with PSCCs similar architecture,but PSCCs are differentiated from both VCs <strong>and</strong>papillomas by <strong>the</strong> presence <strong>of</strong> atypia <strong>of</strong> <strong>the</strong> squamousepi<strong>the</strong>lium covering <strong>the</strong> papillae. The differentiationbetween exophytic <strong>and</strong> papillary SCCs can be more difficultas <strong>the</strong> histologic criteria for <strong>the</strong> diagnosis <strong>of</strong> exophyticSCCs are not clearly defined [30, 355].1.3.5.4 Treatment <strong>and</strong> PrognosisTreatment <strong>of</strong> PSCCs is similar to that <strong>of</strong> conventionalSCCs. Patients with PSCCs are generally believed to havea better prognosis than those with conventional SCCs,although reports in <strong>the</strong> literature are controversial [343,355]. It appears that, because <strong>of</strong> a relatively small number<strong>of</strong> cases published in <strong>the</strong> literature, PSCCs possiblyremain <strong>the</strong> least understood <strong>of</strong> <strong>the</strong> several variants <strong>of</strong>SCC <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck [30].1.3.6 Basaloid Squamous Cell CarcinomaICD-O:8083/3A basaloid squamous cell carcinoma ( BSCC) is a poorlydifferentiated SCC composed <strong>of</strong> basaloid cells <strong>and</strong> squamouscell carcinoma, characterised by an aggressive clinicalcourse. It was first described by Wain et al. in 1986[372]. It has a predilection for <strong>the</strong> upper aerodigestivetract, but also occurs in o<strong>the</strong>r locations such as <strong>the</strong> uterinecervix [140], oesophagus [202], lung [51], <strong>and</strong> anus [90].In <strong>the</strong> upper aerodigestive tract, BSCC shows a predilectionfor <strong>the</strong> hypopharynx (pyriform sinus), base <strong>of</strong><strong>the</strong> tongue, <strong>and</strong> supraglottic larynx [195, 293]; it has alsobeen described in <strong>the</strong> oropharynx [195, 293], oral cavity[69, 72, 159] <strong>and</strong> trachea [277, 312]. The suggested precursor<strong>of</strong> <strong>the</strong> BSCC is a totipotent primitive cell locatedin <strong>the</strong> basal cell layer <strong>of</strong> <strong>the</strong> surface epi<strong>the</strong>lium, or within<strong>the</strong> seromucinous gl<strong>and</strong>s [293, 372].1.3.6.1 AetiologyTobacco <strong>and</strong> alcohol use are strong risk factors for <strong>the</strong>development <strong>of</strong> BSCCs [19].1.3.6.2 Pathologic FeaturesMacroscopically, <strong>the</strong> tumour usually appears as a white,firm, poorly defined, exophytic, polypoid, <strong>and</strong> centrallyulcerated mass with peripheral submucosal infiltration[21].Microscopically, BSCCs are composed <strong>of</strong> small, closelypacked basaloid cells, with hyperchromatic nuclei withor without nucleoli, <strong>and</strong> scant cytoplasm (Fig. 1.17a).The tumour grows in a solid pattern with a lobular configuration,with a frequent peripheral palisading <strong>of</strong> nuclei.Large central necroses <strong>of</strong> <strong>the</strong> comedo type are frequent.Distinctive features <strong>of</strong> BSCCs that are not foundin conventional SCCs are small cystic spaces containingpara aminosalicylate (PAS)- <strong>and</strong> Alcian blue-positivematerial <strong>and</strong> focal stromal hyalinisation [19, 372].BSCCs are always associated with an SCC component,which can present ei<strong>the</strong>r as an in situ or invasiveSCC. The invasive SCC is usually located superficially,<strong>and</strong> is typically well- to moderately differentiated.It may also present as focal squamous differentiationwithin <strong>the</strong> basaloid tumour isl<strong>and</strong>s. The transition between<strong>the</strong> squamous cells <strong>and</strong> <strong>the</strong> basaloid cells is <strong>of</strong>tenabrupt (Fig. 1.17b), or <strong>the</strong>re may be a narrow zone <strong>of</strong>transition.If <strong>the</strong>re is extensive ulceration, only dysplastic changesmay be identifiable in <strong>the</strong> intact surface epi<strong>the</strong>lium[19, 21]. Rarely, BSCCs exhibit a malignant spindle cellcomponent [21, 250]. Metastases may demonstrate basaloidcarcinoma, squamous carcinoma, or both [21].By electron microscopy, desmosomes <strong>and</strong> ton<strong>of</strong>ilamentswere demonstrated in basaloid cells <strong>and</strong> in squamouscells. There were no neurosecretory granules,my<strong>of</strong>ilaments or secretory granules [154, 372].Immunohistochemically, BSCCs express keratin <strong>and</strong>epi<strong>the</strong>lial membrane antigen, but <strong>the</strong> percentage <strong>of</strong> positivecells varies among different reports. It is advised touse a cocktail <strong>of</strong> keratin antibodies (i.e. CAM 5.2, AE-1-AE3) to avoid false-negative results [21]. Some casesexpress carcinoembryonic antigen <strong>and</strong> neuron-specificenolase [19, 195, 318], while expression <strong>of</strong> S-100 protein,vimentin <strong>and</strong> muscle-specific actin varied amongdifferent reports. Vimentin was negative in some studies[69, 195], while Barnes et al. [21] described positivestaining in <strong>the</strong> majority <strong>of</strong> basaloid cells, with a peculiarpattern <strong>of</strong> staining, forming a delicate perinuclear rim.Varying results have also been reported for S-100 immunoreactivity.Some authors described focal immunoreactivityin a few cases [19, 21], while o<strong>the</strong>rs did not findany S-100-positive tumour cells [69, 195, 248]. However,most cases displayed numerous S-100-positive dendriticcells intermingled with <strong>the</strong> tumour cells [9, 21, 195,248]. BSCCs do not express chromogranin, synaptophysin<strong>and</strong> GFAP [19, 21, 195].


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 21Fig. 1.17. Basaloid squamous cell carcinoma.a Closely packed basaloid cells with hyperchromaticnuclei <strong>and</strong> scant cytoplasm,with focal peripheral palisading <strong>of</strong> nuclei.b Abrupt transition between squamous <strong>and</strong>basaloid cells. c Focal squamous differentiationin adenoid cystic carcinoma. Courtesy<strong>of</strong> Dr. Pieter J. Slootwegabc1.3.6.3 Differential DiagnosisDifferential diagnosis includes neuroendocrine carcinoma,adenoid cystic carcinoma, adenocarcinoma <strong>and</strong>adenosquamous carcinoma.Neuroendocrine carcinomas express various neuroendocrinemarkers that help to distinguish neuroendocrinecarcinomas from BSCCs. However, as 60–75%<strong>of</strong> cases <strong>of</strong> BSCC have been reported to express neurone-specificenolase [19, 65, 318] <strong>the</strong> application <strong>of</strong> o<strong>the</strong>rneuroendocrine markers, including chromogranin,CD56, <strong>and</strong> synaptophysin, is advised [19, 65].Adenoid cystic carcinomas, especially <strong>the</strong> solid variant,may resemble BSCCs but adenoid cystic carcinomasrarely show squamous differentiation (Fig. 1.17C). Immunohistochemistrymay also be helpful: tumour cellsin adenoid cystic carcinomas express S-100 protein <strong>and</strong>vimentin, while tumour cells in BSCCs usually do notexpress ei<strong>the</strong>r <strong>of</strong> <strong>the</strong> two markers [21, 195].Adenocarcinomas <strong>and</strong> adenosquamous carcinomascan be distinguished from BSCCs by <strong>the</strong> presence<strong>of</strong> gl<strong>and</strong> formation <strong>and</strong> mucin secretion within <strong>the</strong> tumourcells.1.3.6.4 Treatment <strong>and</strong> PrognosisA BSCC is an aggressive, rapidly growing tumour characterisedby an advanced stage at <strong>the</strong> time <strong>of</strong> diagnosis <strong>and</strong>a poor prognosis. Metastases to <strong>the</strong> regional lymph nodeshave been reported in two-thirds <strong>of</strong> patients [19, 195, 277,293], <strong>and</strong> distant metastases involving lungs, bone, skin<strong>and</strong> brain in 37–50% <strong>of</strong> patients [19, 195, 293].It is generally believed that BSCCs are more aggressivethan conventional SCCs [103, 108, 195, 372, 377].However, some studies indicate that BSCCs exhibit behavioursimilar to that <strong>of</strong> high-grade conventional SCCs<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck [19, 134, 222, 376].The treatment <strong>of</strong> choice is radical surgical excision<strong>and</strong>, because <strong>of</strong> early regional lymph node <strong>and</strong> distantvisceral metastases, radical neck dissection <strong>and</strong> supplementaryradio- <strong>and</strong> chemo<strong>the</strong>rapy [21, 372, 375].


22 N. Gale · N. Zidar1aFig. 1.18. Adenoid squamous cell carcinoma. a Isl<strong>and</strong>s <strong>of</strong> squamouscell carcinoma with pseudogl<strong>and</strong>ular (adenoid) structuresbdue to acantholysis <strong>of</strong> neoplastic cells. b Anastomosing spaces <strong>and</strong>channels mimicking an angiosarcoma.1.3.7 Adenoid Squamous Cell CarcinomaICD-O:8075/3Adenoid squamous cell carcinoma ( adenoid SCC) is anuncommon histopathologic type <strong>of</strong> SCC that was firstrecognised by Lever in 1947 [211]. It resembles an ordinarySCC, but because <strong>of</strong> <strong>the</strong> acantholysis <strong>of</strong> malignantsquamous cells, pseudoluminae are formed, creating<strong>the</strong> appearance <strong>of</strong> gl<strong>and</strong>ular differentiation. There is noevidence <strong>of</strong> true gl<strong>and</strong>ular differentiation or mucin production.Adenoid SCC has been referred to by a variety <strong>of</strong>names such as pseudogl<strong>and</strong>ular SCC, acantholytic SCC,SCC with gl<strong>and</strong>-like features <strong>and</strong> adenoacanthoma.In <strong>the</strong> head <strong>and</strong> neck it arises most frequently in <strong>the</strong>skin (especially in sun-exposed areas) [259, 260], <strong>and</strong>less frequently in mucosal sites <strong>of</strong> <strong>the</strong> upper aerodigestivetract, including <strong>the</strong> lip, oral cavity, tongue <strong>and</strong> nasopharynx[27, 37, 105, 135, 173, 348, 375, 388].1.3.7.1 Pathologic FeaturesAdenoid SCCs are composed <strong>of</strong> isl<strong>and</strong>s <strong>and</strong> cords <strong>of</strong>keratinising SCC; because <strong>of</strong> <strong>the</strong> acantholysis <strong>of</strong> neoplasticcells, pseudogl<strong>and</strong>ular (adenoid) structuresare formed that have central lumina containing detachedacantholytic neoplastic cells, necrotic debris,or <strong>the</strong>y may be empty (Fig. 1.18a). The conventionalsquamous cell carcinoma component is nearly alwayspresent.Acantholysis may lead to <strong>the</strong> formation <strong>of</strong> anastomosingspaces <strong>and</strong> channels, thus mimicking an angiosarcoma(Fig. 1.18b). This variant <strong>of</strong> adenoid SCC istermed pseudovascular adenoid SCC or angiosarcomalikeSCC, <strong>and</strong> has been reported in <strong>the</strong> skin <strong>of</strong> <strong>the</strong> head<strong>and</strong> neck [260], as well as in o<strong>the</strong>r organs, such as breast<strong>and</strong> lungs [18].Immunohistochemically, adenoid SCCs are positivefor epi<strong>the</strong>lial markers, such as cytokeratins <strong>and</strong> epi<strong>the</strong>lialmembrane antigen (EMA); it may also express carcinoembryonicantigen (CEA) <strong>and</strong> vimentin [105].Ultrastructural analysis revealed hemidesmosomes<strong>and</strong> attached ton<strong>of</strong>ilaments, with no gl<strong>and</strong>ular features,thus supporting <strong>the</strong> squamous origin <strong>of</strong> <strong>the</strong> adenoidSCC [388].1.3.7.2 Differential DiagnosisAdenoid SCCs must be differentiated from adenocarcinomas,particularly adenoid cystic carcinomas, adenosquamouscarcinomas, <strong>and</strong> mucoepidermoid carcinomas.This is best achieved by demonstrating that <strong>the</strong>reis no true gl<strong>and</strong> formation <strong>and</strong> that stains for mucin arenegative in adenoid SCCs.Differential diagnosis also includes angiosarcoma,but immunohistochemistry helps to distinguish between<strong>the</strong> two tumours. Angiosarcomas typically expressvascular antigens (CD31, CD34, von Willebr<strong>and</strong>factor) that are negative in adenoid SCCs. Cytokeratin,however, may also be positive in some angiosarcomas[139].1.3.7.3 Treatment <strong>and</strong> PrognosisTreatment <strong>and</strong> prognosis are similar to those for adenoidSCCs <strong>and</strong> conventional SCCs. Some authors, however,believe that adenoid SCCs have aggressive behaviour


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 23<strong>and</strong> a worse prognosis than conventional SCCs [27, 105,348, 388], but <strong>the</strong> number <strong>of</strong> patients reported so far istoo small to draw firm conclusions [328].1.3.8 Adenosquamous CarcinomaICD-O:8560/3Adenosquamous carcinoma (ASC) is a rare malignantepi<strong>the</strong>lial tumour characterised by <strong>the</strong> presence<strong>of</strong> both SCC <strong>and</strong> adenocarcinoma, <strong>and</strong> aggressive behaviour.It occurs in various sites, such as <strong>the</strong> pancreas[68], lung [262], uterine cervix [132], prostate[310], stomach [44] <strong>and</strong> breast [305]. In <strong>the</strong> head <strong>and</strong>neck, it was first reported by Gerughty et al. [131] whodescribed a series <strong>of</strong> 10 patients with nasal, oral <strong>and</strong>laryngeal ASC.Since <strong>the</strong>n, over 150 cases <strong>of</strong> ASC <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck have been reported; <strong>the</strong> most frequent site <strong>of</strong> occurrenceis <strong>the</strong> larynx [8, 83, 124, 192], followed by <strong>the</strong>nose <strong>and</strong> paranasal sinuses [8, 245], oral cavity [8, 192,258, 317, 385], upper lip [234], nasopharynx [234], oropharynx[234] <strong>and</strong> hypopharnyx [83, 234, 313].The histogenesis <strong>of</strong> <strong>the</strong> ASC has not yet been completelyelucidated. Some authors have suggested that itoriginates from <strong>the</strong> salivary <strong>and</strong>/or mucoserous gl<strong>and</strong>s[131] while o<strong>the</strong>rs favour a surface epi<strong>the</strong>lial derivationor a combined gl<strong>and</strong>ular <strong>and</strong> surface epi<strong>the</strong>lial derivation[83]. However, it is becoming increasingly acceptedthat <strong>the</strong> basal cells <strong>of</strong> <strong>the</strong> surface squamous epi<strong>the</strong>lium,which are capable <strong>of</strong> divergent differentiation, are<strong>the</strong> sole origin <strong>of</strong> ASCs [8, 258, 264, 317].1.3.8.1 AetiologyAetiology has not been defined, but cigarette smoking<strong>and</strong> alcohol consumption probably play an importantrole in <strong>the</strong> pathogenesis <strong>of</strong> ASCs, similar to o<strong>the</strong>r types<strong>of</strong> SCC in <strong>the</strong> upper aerodigestive tract [8, 131, 192].1.3.8.2 Pathologic FeaturesAdenosquamous carcinomas do not differ macroscopicallyfrom conventional SCCs. Microscopically, <strong>the</strong>y arecharacterised by <strong>the</strong> presence <strong>of</strong> both adenocarcinoma<strong>and</strong> SCC. The two components occur in close proximity,but are generally distinct <strong>and</strong> separate, <strong>and</strong> are notclosely intermingled as in <strong>the</strong> mucoepidermoid carcinoma.The SCC component can present ei<strong>the</strong>r as an in situor as an invasive SCC, manifesting intercellular bridges,keratin pearl formation or dyskeratosis. The adenocarcinomatouscomponent is usually located in <strong>the</strong> deeperparts <strong>of</strong> <strong>the</strong> tumour; it consists <strong>of</strong> tubular, alveolar orductular structures (Fig. 1.19).Fig. 1.19. Adenosquamous carcinoma: squamous cell carcinomain situ <strong>and</strong> <strong>the</strong> adenocarcinomatous component in <strong>the</strong> deeper part<strong>of</strong> <strong>the</strong> tumour. The two components are in close proximity, thoughseparateThe presence <strong>of</strong> intracytoplasmic mucin can be demonstratedby special techniques, such as PAS, Alcianblue <strong>and</strong> Mayer mucicarmine. Necroses <strong>and</strong> mitoses arecommon [8, 192].Immunohistochemistry has demonstrated positivestaining for cytokeratins with high molecular weightin both <strong>the</strong> SCC <strong>and</strong> <strong>the</strong> adenocarcinomatous components,<strong>and</strong> positive staining for carcinoembryonic antigen(CEA) <strong>and</strong> cytokeratins with low molecular weightin <strong>the</strong> adenocarcinomatous component [8, 234].By electron microscopy, features <strong>of</strong> both squamous<strong>and</strong> adenocarcinomatous differentiation have beendemonstrated [41, 164].1.3.8.3 Differential DiagnosisDifferential diagnosis includes mucoepidermoid carcinoma,adenoid SCC, conventional SCC invading <strong>the</strong>normal salivary gl<strong>and</strong>s, <strong>and</strong> necrotising sialometaplasia.It is important to differentiate ASCs from mucoepidermoidcarcinomas because ASCs have a worse prognosisthan mucoepidermoid carcinomas [131, 192, 313].The histopathologic features favouring <strong>the</strong> diagnosis <strong>of</strong>ASC are: separate <strong>and</strong> distinct areas <strong>of</strong> SCC <strong>and</strong> adeno-


24 N. Gale · N. Zidar1Fig. 1.20. Nasopharyngeal carcinoma.a Isl<strong>and</strong>s <strong>of</strong> poorly differentiated carcinomabeneath <strong>the</strong> surface epi<strong>the</strong>lium, with denselymphocytic infiltration <strong>of</strong> <strong>the</strong> stroma.b In situ hybridisation reveals Epstein-Barrvirus RNA transcripts in <strong>the</strong> nuclei <strong>of</strong> all tumourcellsabcarcinomatous components, <strong>and</strong> <strong>the</strong> involvement <strong>of</strong> <strong>the</strong>surface epi<strong>the</strong>lium exhibiting atypical hyperplasia, carcinomain situ, or invasive SCC.The presence <strong>of</strong> mucin in true gl<strong>and</strong>ular spaces helpsto distinguish ASCs from adenoid SCCs.Conventional SCCs invading or entrapping <strong>the</strong> normalsalivary or mucoserous gl<strong>and</strong>s can be confused withASC, especially in small biopsy specimens. In such cases,preservation <strong>of</strong> lobular gl<strong>and</strong> architecture <strong>and</strong> lack <strong>of</strong>significant atypia are observed, helping to distinguishconventional SCCs from ASCs.Finally, ASCs must be differentiated from necrotisingsialometaplasia, which is a benign condition. The histopathologicfeatures suggesting <strong>the</strong> diagnosis <strong>of</strong> necrotisingsialometaplasia are: surface ulceration, localisationin minor salivary gl<strong>and</strong>s, lobular architecture, partialnecrosis <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>, <strong>and</strong> squamous metaplasia<strong>of</strong> <strong>the</strong> salivary ducts.ICD-O:8082/3Lymphoepi<strong>the</strong>lial carcinoma (LEC) is a poorly differentiatedSCC or undifferentiated carcinoma, associatedwith dense lymphocytic stromal infiltration. It ismorphologically indistinguishable from nasopharyngealcarcinoma type 3 (WHO classification) [381]. Itwas originally described in <strong>the</strong> nasopharynx in 1921by Regaud <strong>and</strong> Reverchon [294], <strong>and</strong> independently bySchmincke [315]. Synonyms for LEC include lymphoepi<strong>the</strong>lioma,nasopharyngeal-type carcinoma, Regaud<strong>and</strong> Schmincke-type lymphoepi<strong>the</strong>lioma, <strong>and</strong> undifferentiatedcarcinoma. The specific features <strong>of</strong> <strong>the</strong> nasopharyngealcarcinoma are extensively discussed inChap. 6.Apart from <strong>the</strong> nasopharynx, it rarely occurs in o<strong>the</strong>rlocations in <strong>the</strong> head <strong>and</strong> neck, such as <strong>the</strong> oropharynx,salivary gl<strong>and</strong>s, tonsils, tongue, s<strong>of</strong>t palate, uvula,floor <strong>of</strong> <strong>the</strong> mouth, sinonasal tract, larynx <strong>and</strong> hypopharynx[67, 93, 120, 227, 392], as well as elsewherein <strong>the</strong> body including <strong>the</strong> lung, urinary bladder, uterinecervix, breast, skin <strong>and</strong> stomach [113].1.3.8.4 Treatment <strong>and</strong> PrognosisThe ASC has a more aggressive course than <strong>the</strong> conventionalSCC [131, 258, 313], with a tendency toward earlylymph node metastases, frequent local recurrences, <strong>and</strong>occasional dissemination [192]. The reported 5-yearsurvival rate is between 13 <strong>and</strong> 25% [124, 131, 192].The treatment <strong>of</strong> choice is radical surgical excision.Irradiation alone has had poor results [124, 192, 313].Some reports indicate that radical surgery combinedwith irradiation may improve <strong>the</strong> survival rate [6].1.3.9.1 AetiologyNasopharyngeal carcinomas are aetiopathogeneticallyassociated with <strong>the</strong> Epstein-Barr virus (EBV; Fig. 1.20b)[265]. Apart from nasopharyngeal carcinomas, EBV hasbeen also implicated in <strong>the</strong> pathogenesis <strong>of</strong> LECs <strong>of</strong> <strong>the</strong>salivary gl<strong>and</strong>s, as well as undifferentiated carcinomas<strong>of</strong> <strong>the</strong> stomach, lung <strong>and</strong> thymus [153, 160]. In contrast,it seems that EBV plays little, if any role in <strong>the</strong> pathogenesis<strong>of</strong> LEC in o<strong>the</strong>r locations in <strong>the</strong> head <strong>and</strong> neck [93,153, 227, 392].1.3.9 Lymphoepi<strong>the</strong>lial Carcinoma1.3.9.2 Pathologic FeaturesThe LEC is composed <strong>of</strong> small clusters or aggregates(Schmincke pattern) or large syncytial masses (Regaudpattern) <strong>of</strong> cells. Tumour cells have oval or round ve-


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 25sicular nuclei, <strong>and</strong> one to three prominent nucleoli(Fig. 1.20a). The cytoplasm is sparse <strong>and</strong> poorly defined.Normal <strong>and</strong> abnormal mitoses may be numerous. Necrosesmay be present.The LEC may exist in two histological forms: asa pure LEC <strong>and</strong> as a mixed form, composed <strong>of</strong> bothLEC <strong>and</strong> conventional SCC; such a mixture has beenobserved both in primary <strong>and</strong> metastatic tumours[227].The stroma in LECs is densely infiltrated by T lymphocytes;stromal inflammatory infiltration may alsocontain plasma cells, follicular dendritic cells <strong>and</strong> eosinophils.1.3.9.3 Differential DiagnosisDifferential diagnosis includes malignant lymphoma,in particular diffuse large B-cell lymphoma, as well asmalignant melanoma <strong>and</strong> rhabdomyosarcoma. Differentiationis achieved by <strong>the</strong> use <strong>of</strong> appropriate immunohistochemicalstaining. The vast majority <strong>of</strong>LECs are positive for cytokeratin <strong>and</strong> negative for leukocytecommon antigen as well as o<strong>the</strong>r lymphocyteantigens. Cytokeratin positivity has been reported inrare lymphomas [123], but leukocyte common antigenpositivity in <strong>the</strong> tumour cells <strong>of</strong> LECs has not yet beenreported. A negative reaction to S-100, HMB-45 <strong>and</strong>melan-A helps to differentiate LECs from malignantmelanomas.1.3.9.4 Treatment <strong>and</strong> PrognosisLymphoepi<strong>the</strong>lial carcinomas are more aggressive thanconventional SCCs, with a higher incidence <strong>of</strong> cervicallymph node metastases, <strong>and</strong> a propensity for distant metastases,mostly to <strong>the</strong> lung, liver <strong>and</strong> bones [93, 227]. Ina series <strong>of</strong> 34 patients with LECs, 76% <strong>of</strong> patients hadlymph node metastases at <strong>the</strong> time <strong>of</strong> diagnosis, <strong>and</strong>36% had distant metastases [93].The LEC is a radiosensitive tumour <strong>and</strong> radio<strong>the</strong>rapyis an appropriate initial <strong>the</strong>rapy for patients withLECs. Surgical treatment should be reserved for patientswith persistent disease after completing radio<strong>the</strong>rapy.In those patients adjuvant chemo<strong>the</strong>rapy is also recommendedin an attempt to decrease <strong>the</strong> rate <strong>of</strong> distant metastases[93].1.4 Second Primary TumoursPatients with SCCs <strong>of</strong> <strong>the</strong> upper aerodigestive tract areat high risk <strong>of</strong> developing a second primary tumour(SPT) at a separate anatomic site from <strong>the</strong> index (first)tumour. The SPT is synchronous if it is diagnosedwithin 6 months after <strong>the</strong> index tumour, or metachronousif it is diagnosed more than 6 months after <strong>the</strong>index tumour. Synchronous tumours are simultaneousif <strong>the</strong>y are discovered at <strong>the</strong> same time as <strong>the</strong> index tumour.The median prevalence <strong>of</strong> SPT in patients with an indextumour in <strong>the</strong> upper aerodigestive tract is 9% [149,291, 337]. The site <strong>of</strong> <strong>the</strong> SPT is affected by <strong>the</strong> site <strong>of</strong><strong>the</strong> index tumour. In patients with an index tumour in<strong>the</strong> oral cavity, pharynx <strong>and</strong> oesophagus, <strong>the</strong> SPT tendsto arise in <strong>the</strong> same location. In patients with index tumoursin <strong>the</strong> larynx, <strong>the</strong> SPT tends to be located in <strong>the</strong>lungs [174].The risk <strong>of</strong> developing an SPT closely correlates with<strong>the</strong> use <strong>of</strong> tobacco <strong>and</strong> alcohol abuse, <strong>and</strong> is more th<strong>and</strong>oubled in patients who smoke <strong>and</strong> drink comparedwith those who do not smoke <strong>and</strong> drink [207]. Moreover,<strong>the</strong>re is a direct dose-dependent relationship betweentobacco <strong>and</strong> alcohol exposure <strong>and</strong> <strong>the</strong> risk <strong>of</strong> SPT.It is now accepted that long-term exposure to tobacco<strong>and</strong>/or alcohol causes extensive <strong>and</strong> diffuse DNA changesleading to widespread genetic damage or “field cancerisation”<strong>of</strong> <strong>the</strong> whole respiratory tract <strong>and</strong> <strong>the</strong> upper digestivetract [324].The prognosis for patients with SPTs is poor, beingworse for synchronous SPTs than for metachronous tumours.They generally present with a more advanced Tstage, <strong>and</strong> have a much lower 5-year survival than <strong>the</strong>index tumour [291].It is <strong>the</strong>refore imperative for a panendoscopy to beperformed at <strong>the</strong> time <strong>of</strong> diagnosis <strong>of</strong> <strong>the</strong> index tumour,not only as a part <strong>of</strong> <strong>the</strong> staging procedure, but also tolook for an SPT [291].1.5 Tumour Spread <strong>and</strong> MetastasisSquamous cell carcinomas may spread directly to contiguousstructures, as well as via lymphatic <strong>and</strong> bloodvessels, giving rise to regional lymph node <strong>and</strong> distantmetastases, or metastases along <strong>the</strong> nerves. The behaviour<strong>and</strong> spread <strong>of</strong> SCCs are affected by various factors,<strong>the</strong> most important being <strong>the</strong> site <strong>of</strong> <strong>the</strong> primarytumour. This has been attributed to <strong>the</strong> rich vascular<strong>and</strong> lymphatic network in certain areas, such as <strong>the</strong>base <strong>of</strong> <strong>the</strong> tongue, where metastatic rates are significantlyhigher than for similar-sized tumours on <strong>the</strong>oral tongue. Similarly, poorly vascularised areas, suchas <strong>the</strong> glottis, are associated with a lower rate <strong>of</strong> metastases[106].O<strong>the</strong>r factors important for <strong>the</strong> spread <strong>and</strong> behaviour<strong>of</strong> SCCs include <strong>the</strong> size <strong>and</strong> <strong>the</strong> differentiation<strong>of</strong> <strong>the</strong> tumour, as well as poorly defined factors <strong>of</strong> <strong>the</strong>host, i.e. <strong>the</strong> immune status <strong>and</strong> genetic susceptibility[130].


26 N. Gale · N. Zidar1associated with an increased risk <strong>of</strong> local recurrence, regionallymph node metastases <strong>and</strong> decreased survival[101, 233, 333, 383].1.5.3 Regional Lymph Node MetastasesFig. 1.21. Tumour emboli in a lymph vessel <strong>and</strong> vein1.5.1 Invasion <strong>of</strong> Lymphatic<strong>and</strong> Blood VesselsExperimental studies have shown that metastatic progressionis initiated by local invasion: select tumourcells are released from <strong>the</strong> tumour where <strong>the</strong>y gain entryto <strong>the</strong> lymphatic system or circulation, mainly via <strong>the</strong>production <strong>of</strong> tumour-derived proteolytic enzymes <strong>and</strong>angiogenic factors [220].Cancer cells commonly invade thin-walled lymphaticvessels, capillaries <strong>and</strong> veins (Fig. 1.21), whereas thickerwalledarterioles <strong>and</strong> arteries are relatively resistant. Theappearance <strong>of</strong> vascular invasion should not be consideredsynonymous with metastasis, because most <strong>of</strong> <strong>the</strong>tumour cells that enter <strong>the</strong> lymphatic system <strong>and</strong> circulationare destroyed [1]. However, <strong>the</strong> penetration <strong>of</strong> tumourcells in <strong>the</strong> lymphatic <strong>and</strong> blood vessels is associatedwith a high probability <strong>of</strong> regional lymph node<strong>and</strong> distant metastases. Fur<strong>the</strong>rmore, it allows <strong>the</strong> tumourto spread beyond <strong>the</strong> apparent margins. The presence<strong>of</strong> vascular invasion is <strong>the</strong>refore associated withan increased incidence <strong>of</strong> recurrence <strong>and</strong> poor survival[383].1.5.2 Perineural InvasionIn perineural invasion, <strong>the</strong> tumour cells enter <strong>the</strong> perineuralspace <strong>and</strong> spread both proximally <strong>and</strong> distallyalong <strong>the</strong> nerve fibre. Even though a perineural spread<strong>of</strong> more than 2 cm is unusual, <strong>the</strong> travelling <strong>of</strong> tumourcells up to 12 cm away from <strong>the</strong> primary tumour sitealong <strong>the</strong> perineural space has been described [101,370].Patients with perineural invasion may be asymptomatic,or may experience pain <strong>and</strong> pares<strong>the</strong>sia [40]. It appearsthat perineural invasion is a poor prognostic sign,Squamous cell carcinomas <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck havea high tendency to metastasise to <strong>the</strong> regional lymphnodes. The localisation <strong>and</strong> frequency <strong>of</strong> <strong>the</strong> lymph nodemetastases depend upon <strong>the</strong> site <strong>and</strong> size <strong>of</strong> <strong>the</strong> primarytumour. Large metastases can be detected clinically byexamination or using ultrasound or radiographic methods.Smaller metastases evade clinical detection, but aredetected by light microscopy [111].Routine analysis <strong>of</strong> neck dissection specimens is usuallylimited to <strong>the</strong> examination <strong>of</strong> a few sections <strong>of</strong> eachnode stained by haematoxylin-eosin. During such routineanalysis, small metastases can easily be missed. Ithas been demonstrated that with more sensitive techniques,nodal metastases can be detected in 8–20% <strong>of</strong>patients in whom metastases had not been found duringroutine histologic examination [11, 146, 147]. The mostcommonly used sensitive techniques for <strong>the</strong> detection <strong>of</strong>small metastases are serial section light microscopy, immunohistochemistry<strong>and</strong> molecular analysis [130, 273,307, 379].The prognostic significance <strong>of</strong> lymph node metastaseshas been extensively studied. Metastasis in <strong>the</strong>lymph nodes is <strong>the</strong> most significant adverse prognosticfactor in head <strong>and</strong> neck SCCs. The 5-year survival is decreasedby approximately 50% in patients with lymphnode metastases compared with patients without nodalinvolvement [10, 25, 320]. The number <strong>and</strong> size <strong>of</strong> positivenodes, <strong>the</strong>ir level in <strong>the</strong> neck <strong>and</strong> <strong>the</strong> presence <strong>of</strong>extracapsular spread are <strong>the</strong> most important prognosticparameters for nodal status [10, 85, 109, 230].1.5.3.1 Extracapsular Spreadin Lymph Node MetastasesCancer cells initially lodge in <strong>the</strong> marginal sinus, <strong>and</strong><strong>the</strong>n extend throughout <strong>the</strong> lymph node. Metastasesmay be confined to <strong>the</strong> lymph node, or may penetrate <strong>the</strong>capsule <strong>and</strong> infiltrate <strong>the</strong> perinodal tissue; this pattern<strong>of</strong> growth has been referred to as extracapsular spread(ECS). Extracapsular spread is fur<strong>the</strong>r divided into macroscopic<strong>and</strong> microscopic ECS [62]; macroscopic ECS isevident to <strong>the</strong> naked eye during <strong>the</strong> laboratory dissection<strong>of</strong> <strong>the</strong> surgical specimen <strong>and</strong> is later confirmed byhistological assessment. It usually involves not only <strong>the</strong>perinodal fibro-adipose tissue, but also <strong>the</strong> surroundingstructures. Microscopic ECS is only evident on histologicexamination <strong>and</strong> is usually limited to <strong>the</strong> adjacentperinodal fibro-adipose tissue.


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 27Extracapsular spread is a significant predictor <strong>of</strong>both regional recurrence <strong>and</strong> <strong>the</strong> development <strong>of</strong> distantmetastases resulting in decreased survival [109, 155, 175,335, 337]. In some studies, ECS has been shown as a betterpredictor than <strong>the</strong> resection margins. It has <strong>the</strong>reforebeen suggested that ECS should be incorporated into <strong>the</strong>staging system for surgically managed patients [380].Some studies, on <strong>the</strong> contrary, have not confirmed <strong>the</strong>independent prognostic significance <strong>of</strong> extracapsularspread [230, 280].1.5.3.2 Metastases in <strong>the</strong> S<strong>of</strong>t Tissue<strong>of</strong> <strong>the</strong> <strong>Neck</strong>In some patients, an SCC in <strong>the</strong> s<strong>of</strong>t tissue <strong>of</strong> <strong>the</strong> neck isfound, with no evidence <strong>of</strong> lymph nodes being present.These s<strong>of</strong>t tissue metastases may be <strong>the</strong> result <strong>of</strong> ei<strong>the</strong>rtotal effacement <strong>of</strong> a lymph node by <strong>the</strong> SCC, or extralymphaticspread <strong>of</strong> <strong>the</strong> SCC [176].It has been shown that <strong>the</strong> presence <strong>of</strong> s<strong>of</strong>t tissue metastasesis associated with an aggressive clinical course<strong>and</strong> poor survival [176, 368]. In a study <strong>of</strong> 155 patients,survival was significantly shorter for patients with s<strong>of</strong>ttissue metastases than those without nodal metastases<strong>and</strong> those with nodal metastases without extracapsularspread; it was similar to that for patients with lymphnode metastases with extracapsular spread [176].1.5.4 Distant MetastasisDistant metastases in patients with head <strong>and</strong> neck cancerare usually defined as metastases below <strong>the</strong> clavicle,<strong>and</strong> may be <strong>the</strong> result <strong>of</strong> lymphogenic or haematogenousspread. Lymphogenic spread results in distant lymphnode metastases; <strong>the</strong> most commonly affected distantnodes are <strong>the</strong> mediastinal, axillary <strong>and</strong> inguinal nodes[7]. Haematogenous spread results in distant metastases,most commonly to <strong>the</strong> lung, liver <strong>and</strong> bones, followed by<strong>the</strong> skin <strong>and</strong> brain [84, 157, 198, 208, 337, 362, 387]. Metastaseshave been also described in <strong>the</strong> small intestine[384], spleen [3] <strong>and</strong> <strong>the</strong> cavernous sinus [362].Distant metastases in head <strong>and</strong> neck SCCs are infrequent,but may occur in <strong>the</strong> late stages <strong>of</strong> <strong>the</strong> disease,with <strong>the</strong> reported incidence between 3 <strong>and</strong> 8.5%[337, 387]. Postmortem studies have shown a higher incidence<strong>of</strong> distant metastases, ranging from 24 to 57%[266, 325, 393].The incidence <strong>of</strong> distant metastases depends on <strong>the</strong>site <strong>of</strong> <strong>the</strong> primary tumour, as well as <strong>the</strong> initial size <strong>of</strong><strong>the</strong> tumour <strong>and</strong> <strong>the</strong> presence <strong>of</strong> nodal metastases [59,198, 253]. The highest incidence <strong>of</strong> distant metastaseshas been reported in hypopharyngeal SCCs, followed by<strong>the</strong> SCCs <strong>of</strong> <strong>the</strong> tongue [198].Most distant metastases become clinically apparent2 years after diagnosis <strong>of</strong> <strong>the</strong> initial tumour. The averagesurvival once distant metastases are diagnosed rangesbetween 4 <strong>and</strong> 7 months [208].1.5.5 MicrometastasisMicrometastasis is defined as a microscopic deposit <strong>of</strong>malignant cells, smaller than 2–3 mm, that are segregatedspatially from <strong>the</strong> primary tumour [193]. The fate<strong>of</strong> micrometastases is uncertain; <strong>the</strong> majority <strong>of</strong> <strong>the</strong>mare probably destined for destruction or dormancy, <strong>and</strong>only a small percentage <strong>of</strong> circulating tumour cells survive<strong>and</strong> initiate a metastatic focus [1].The fundamental characteristic <strong>of</strong> micrometastasisis <strong>the</strong> absence <strong>of</strong> a specific blood supply. Micrometastasesare thus dependent on passive diffusion for oxygen<strong>and</strong> nutrient supply. Experimental studies haveshown that without new blood vessel formation (neoangiogenesis),<strong>the</strong> growth <strong>of</strong> tumour cells is limited to2–3 mm <strong>and</strong> may remain dormant for months or evenyears. During dormancy, <strong>the</strong> proliferation is balancedby an equivalent rate <strong>of</strong> cell death by apoptosis. Afterinduction <strong>of</strong> neoangiogenesis, apoptosis is significantlyreduced, but <strong>the</strong> proliferation rate remains unchanged,<strong>and</strong> <strong>the</strong> growth <strong>of</strong> <strong>the</strong> clinically overt metastasiscan occur because <strong>of</strong> <strong>the</strong> increased survival <strong>of</strong> <strong>the</strong>tumour cells [158].Micrometastases can be detected anywhere in <strong>the</strong>body, but most frequently in <strong>the</strong> lymph nodes, in <strong>the</strong>surgical margins, in <strong>the</strong> blood <strong>and</strong> in bone marrow[112]. Their detection can be accomplished by serial sectioninglight microscopy, immunohistochemistry, <strong>and</strong>/or molecular analysis [35, 112, 130, 146].The clinical <strong>and</strong> prognostic implication <strong>of</strong> micrometastasesis still uncertain. It has been suggested that residualmicrometastatic tumour cells may increase <strong>the</strong>risk <strong>of</strong> tumour recurrence, thus resulting in failure <strong>of</strong><strong>the</strong> primary treatment. Fur<strong>the</strong>rmore, <strong>the</strong> presence <strong>of</strong> tumourcells in <strong>the</strong> blood <strong>and</strong>/or bone marrow may be anindicator <strong>of</strong> a generalised disease with possible disseminationto many organs [163]. Several studies have demonstratedthat lymph node micrometastases are associatedwith a high risk <strong>of</strong> recurrence <strong>and</strong> poor survivalin patients with carcinoma <strong>of</strong> <strong>the</strong> breast, oesophagus,stomach, colon <strong>and</strong> lung [163], but few studies have beenfocused on <strong>the</strong> clinical significance <strong>of</strong> micrometastasesin SCCs <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck [112, 379].It appears that <strong>the</strong> detection <strong>of</strong> micrometastases isa promising approach that might enable us to identifyc<strong>and</strong>idates for adjuvant treatment strategies [163]. However,fur<strong>the</strong>r studies are needed to define more precisely<strong>the</strong> clinical implication <strong>of</strong> micrometastases, as well as<strong>the</strong> most appropriate method for <strong>the</strong>ir detection.


28 N. Gale · N. Zidar11.6 Molecular <strong>Pathology</strong><strong>of</strong> Squamous Cell CarcinomaMalignant tumours arise clonally from transformedcells that have undergone specific genetic alterations intumour suppressor genes <strong>and</strong> proto-oncogenes [117], aswell as telomerase re-activation [225, 226, 256]. Loss <strong>of</strong>chromosomal region 9p21 is <strong>the</strong> most common geneticchange in head <strong>and</strong> neck carcinogenesis with consequentinactivation <strong>of</strong> <strong>the</strong> p16 gene [168, 169]. A frequentevent is also mutation <strong>of</strong> <strong>the</strong> p53 gene located at 17p13;it occurs in approximately 50% <strong>of</strong> patients with SCCs<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck [255, 257, 270]. Loss <strong>of</strong> retinoblastomagene (Rb1) expression is seen in less than 20% <strong>of</strong>cases, although LOH at 13q14 is present in 60% or more<strong>of</strong> SCCs, suggesting <strong>the</strong> existence <strong>of</strong> (an)o<strong>the</strong>r tumoursuppressor gene(s) neighbouring Rb1 [256].The activation <strong>of</strong> oncogenes also occurs, such as cyclinD1 amplification, which has been described in one-third<strong>of</strong> patients with SCCs <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck, <strong>and</strong> is associatedwith advanced disease [167, 272]. Amplification <strong>of</strong>o<strong>the</strong>r oncogenes, e.g. c-myc <strong>and</strong> epidermal growth factorreceptor, has also been described in 6–25% <strong>of</strong> patientswith SCCs <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck [119, 121, 269], while rasmutations probably do not play a significant role in <strong>the</strong>development <strong>of</strong> head <strong>and</strong> neck SCCs [119].Molecular pathology has significantly deepened ourinsight into genetic alterations occurring in <strong>and</strong> beingprobably responsible for cancer development. Moreover,it <strong>of</strong>fers <strong>the</strong> opportunity for tissue characterisation, i.e.detection <strong>of</strong> tumour cells, distinction between multipleprimary tumours <strong>and</strong> metastatic disease, <strong>and</strong> <strong>the</strong> risk <strong>of</strong>progression <strong>of</strong> premalignant lesions, going beyond morphologicaltechniques that have been used in pathologyuntil now. The question remains, however, whe<strong>the</strong>r<strong>the</strong>re is any practical impact <strong>of</strong> <strong>the</strong>se new techniques regardingdiagnosis, prognosis <strong>and</strong> management.1.6.1 Detecting Tumour CellsThe introduction <strong>of</strong> methods more sensitive than histologyfor detecting tumour cells in surgical margins <strong>and</strong>lymph nodes could be helpful in obtaining better treatmentresults for patients with SCCs <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck[53, 261, 361]. Their application, however, requires somecritical remarks.First, if histology is inadequate for reliable margin assessment,a lot <strong>of</strong> cases <strong>of</strong> SCC <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck thathave been classified as tumour-free after surgery by thismethod should never<strong>the</strong>less show recurrence at <strong>the</strong> sitefrom which <strong>the</strong> tumour was removed.Secondly, <strong>the</strong> usefulness <strong>of</strong> molecular detection <strong>of</strong> tumourcells in lymph nodes should be demonstrated bytransferring patients from <strong>the</strong> histopathologically assessedN0 stage to <strong>the</strong> N+ stage. Merely detecting additionalpositive nodes in patients already classified as N+is <strong>of</strong> debatable value.In <strong>the</strong> third place, genetically altered cells are not alwaystumour cells. The whole epi<strong>the</strong>lial lining <strong>of</strong> <strong>the</strong> upperaerodigestive tract bears <strong>the</strong> genetic burden <strong>of</strong> <strong>the</strong>carcinogenetic agents that cause SCCs <strong>and</strong>, <strong>the</strong>refore,<strong>the</strong>se cells could have arisen independently from <strong>the</strong> invasivetumour [50, 268].Also, <strong>the</strong>re is no uniformity as to what constitutesa positive or negative surgical margin [26]. There is abroad spectrum <strong>of</strong> histological appearance betweennormal epi<strong>the</strong>lium <strong>and</strong> fully developed SCC. If geneticallyaltered cells are found in areas free <strong>of</strong> tumour butwith atypical hyperplasia (severe dysplasia), molecularpathology does not provide any information comparedwith conventional microscopical examination.In a recently published study, <strong>the</strong> value <strong>of</strong> molecularpathology was compared with traditional histology in<strong>the</strong> assessment <strong>of</strong> surgical margins <strong>and</strong> neck nodes in patientswith SCCs <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck [326]. It was foundthat from patients with microscopically positive margins,22% had recurrence at <strong>the</strong> primary site. In contrast, <strong>of</strong> <strong>the</strong>cases with histologically tumour-free margins, only 4%showed recurrence <strong>of</strong> <strong>the</strong> tumour at <strong>the</strong> primary site. Theauthors concluded that conventional histology adequatelyidentifies patients with SCCs <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck atrisk <strong>of</strong> local recurrence, leaving only very limited roomfor improvement using more sophisticated methods.Regarding <strong>the</strong> neck, local recurrence was observedin 12 out <strong>of</strong> 107 cases in which a previous neck dissectionwas reported to be tumour-free. These neck recurrencescould be due to <strong>the</strong> presence <strong>of</strong> micrometastasesnot detected by microscopy <strong>and</strong> improved methods fordetecting <strong>the</strong>m may reduce this number. However, it isstill uncertain whe<strong>the</strong>r micrometastatic disease has <strong>the</strong>same clinical significance as metastatic disease detectedby conventional methods [104].The authors concluded that <strong>the</strong> added clinical value<strong>of</strong> molecular pathology over histology in detecting tumourcells in surgical margins in SCCs <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck does not justify <strong>the</strong> effort. For lymph nodes, suchan added value is still under discussion [326].1.6.2 Clonal AnalysisPatients with SCCs <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck are at risk <strong>of</strong> <strong>the</strong>development <strong>of</strong> multiple primary SCCs, not only in <strong>the</strong>head <strong>and</strong> neck region, but also in <strong>the</strong> lung. Therefore, itis not always clear whe<strong>the</strong>r a patient who has multiplelesions ei<strong>the</strong>r synchronically or metachronically suffersfrom one single disseminated disease or from multipleprimary cancers. If all tumour deposits show SCC, histologycannot distinguish between both possibilitieswhereas both situations require different treatment approaches.If histologically similar tumours differ geneti-


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 29cally, molecular pathology could be decisive in making<strong>the</strong> distinction. Such a clonal marker should be differentin different tumours <strong>and</strong> not subject to alterations duringtumour progression <strong>and</strong> metastasis. The p53 genemeets <strong>the</strong>se requirements; o<strong>the</strong>r markers such as loss<strong>of</strong> heterozygosity analysis are less stable [271, 347, 358,359]. Only in tumours lying close to each o<strong>the</strong>r p53 mutationanalysis may be unreliable as separate tumoursdeveloping within a single neoplastic field may have <strong>the</strong>same p53 mutation [50]. This, however, does not detractfrom <strong>the</strong> main value <strong>of</strong> p53 gene analysis as a tool to distinguishbetween lung lesions as distant metastasis fromSCCs <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck or second primary cancer in<strong>the</strong> lung.1.6.3 Assessment <strong>of</strong> Risk<strong>of</strong> Malignant ProgressionSquamous cell carcinomas <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck may bepreceded by premalignant alterations showing epi<strong>the</strong>lialchanges <strong>of</strong> varying degrees. Molecular pathology hasbeen shown to be helpful in identifying lesions at risk<strong>of</strong> fur<strong>the</strong>r malignant progression. Chromosomal lossesat loci 3p <strong>and</strong> 9p have been shown to occur in mucosallesions that subsequently turned into cancer, but as notall <strong>of</strong> <strong>the</strong> lesions with <strong>the</strong>se genetic alterations progress,o<strong>the</strong>r changes also play a role: additional chromosomallosses, chromosomal polysomies <strong>and</strong> p53 protein expression[204, 306]. Also, suprabasal expression <strong>of</strong> p53protein <strong>and</strong> <strong>the</strong> presence <strong>of</strong> cells with abnormal DNAcontent predict malignant transformation, even in <strong>the</strong>absence <strong>of</strong> overt morphologic epi<strong>the</strong>lial alterations [82,344].1.6.4 DNA/RNA Pr<strong>of</strong>ilingin Predicting Metastatic DiseaseDNA alterations <strong>and</strong> RNA expression pr<strong>of</strong>iles could beuseful in distinguishing between patients with SCCs <strong>of</strong><strong>the</strong> head <strong>and</strong> neck with <strong>and</strong> without lymph node metastasis,thus diminishing <strong>the</strong> number <strong>of</strong> elective neck dissectionsin N0 patients. Data on this issue are just beginningto appear. Their application in patient managementlies in <strong>the</strong> future [81, 148].References1. Abati A, Liotta LA (1996) Looking forward in diagnostic pathology.The molecular superhighway. Cancer 78:1–32. Abbey LM, Page DG, Sawyer DR (1980) The clinical <strong>and</strong> histopathologicfeatures <strong>of</strong> a series <strong>of</strong> 464 oral squamous cellpapillomas. Oral Surg Oral Med Oral Pathol 49:419–4283. Abramson AL, Parisier SC, Zamansky MJ, Sulka M (1971)Distant metastases from carcinoma <strong>of</strong> <strong>the</strong> larynx. Laryngoscope81:1503–15114. Abramson AL, Steinberg BM, Winkler B (1987) Laryngealpapillomatosis: clinical, histopathologic <strong>and</strong> molecular studies.Laryngoscope 97:678–6855. Ackerman LV (1948) Verrucous carcinoma <strong>of</strong> <strong>the</strong> oral cavity.Surgery 23:670–6786. Aden KK, Niehans G, Adams GL, Abdel-Fattah HM (1988)Adenosquamous carcinoma <strong>of</strong> <strong>the</strong> larynx <strong>and</strong> hypopharynxwith five new case presentations. Trans Am Laryngol Assoc109:216–2217. Alavi S, Namazie A, Sercarz JA, Wang MB, Blackwell KE(1999) Distant lymphatic metastasis from head <strong>and</strong> neck cancer.Ann Otol Laryngol 108:860–8638. Alos L, Castillo M, Nadal A, Caballero M, Mall<strong>of</strong>re C, PalacinA, Cardesa A (2004) Adenosquamous carcinoma <strong>of</strong> <strong>the</strong>head <strong>and</strong> neck: criteria for diagnosis in a study <strong>of</strong> 12 cases.Histopathology 44:1–109. Altavilla G, Mannara GM, Rinaldo A, Ferlito A (1999) Basaloidsquamous cell carcinoma <strong>of</strong> oral cavity <strong>and</strong> oropharynx.ORL J Otorhinolaryngol Relat Spec 61:169–17310. Alvi A, Johnson JT (1996) Extracapsular spread in <strong>the</strong> clinicallynegative neck (N0): implications <strong>and</strong> outcome. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 114:65–7011. Ambrosch P, Bricbeck U (1996) Detection <strong>of</strong> nodal micrometastasisin head <strong>and</strong> neck cancer by serial sectioning <strong>and</strong> immunostaining.Oncology 10:1221–122612. Ambrosch P, Schlott T, Hilmes D, Ruschenburg I (2001) p16alterations <strong>and</strong> retinoblastoma protein expression in squamouscell carcinoma <strong>and</strong> neighboring dysplasia from <strong>the</strong> upperaerodigestive tract. Virchows Arch 438:343–34913. Ansari-Lari MA, Hoque MO, Califano J, Westra WH (2002)Immunohistochemical p53 expression patterns in sarcomatoidcarcinomas <strong>of</strong> <strong>the</strong> upper respiratory tract. Am J SurgPathol 26:1024–103114. Axell T, Pindborg JJ, Smith CJ, van der Waal I (1996) Oralwhite lesions with special reference to precancerous <strong>and</strong> tobacco-relatedlesions: conclusions <strong>of</strong> an international symposiumheld in Uppsala, Sweden, May 18–21 1994. InternationalCollaborative Group on Oral White Lesions. J OralPathol Med 25:49–5415. Azorin D, Rodriguez-Peralto JL, Garcia-Garcia E, SalamancaJ (2003) Cutaneous papillary squamous cell carcinoma.Report <strong>of</strong> three new cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. VirchowsArch 442:298–30216. Azzimonti B, Hertel L, Aluffi P, Pia F, Monga G, Zocchi M,L<strong>and</strong>olfo S, Gariglio M (1999) Demonstration <strong>of</strong> multipleHPV types in laryngeal premalignant lesions using polymerasechain reaction <strong>and</strong> immunohistochemistry. J MedVirol 59:110–11617. Balažic J, Mašera A, Poljak M (1997) Sudden death caused bylaryngeal papillomatosis. Acta Otolaryngol 527:111–11318. Banerjee SS, Eyden BP, Wells S, McWilliam LJ, Harris M(1992) Pseudoangiosarcomatous carcinoma – a clinicopathologicalstudy <strong>of</strong> 7 cases. Histopathology 21:13–2319. Banks ER, Frierson HF, Mills SE, George E, Zarbo RJ, SwansonPE (1992) Basaloid squamous cell carcinoma <strong>of</strong> <strong>the</strong> head<strong>and</strong> neck. Am J Surg Pathol 16:939–94620. Barnes L (2001) Diseases <strong>of</strong> <strong>the</strong> larynx, hypopharynx, <strong>and</strong>esophagus. In: Barnes’ surgical pathology <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck, 2nd edn. Dekker, New York, pp 128–23721. Barnes L, Ferlito A, Altavilla G, MacMillan C, Rinaldo A,Doglioni C (1996) Basaloid squamous cell carcinoma <strong>of</strong> <strong>the</strong>head <strong>and</strong> neck: clinicopathologic features <strong>and</strong> differential diagnosis.Ann Otol Rhinol Laryngol 105:75–8222. Barnes L, Yunis EJ, Krebs FJ III, Sonmez-Alpan E (1991) Verrucavulgaris <strong>of</strong> <strong>the</strong> larynx. Demonstration <strong>of</strong> human papillomavirustypes 6/11 by in situ hybridization. Arch PatholLab Med 115:895–89923. Barona de Guzman R, Martorell MA, Basterra J, ArmengotM, Montoro A, Montoro J (1993) Analysis <strong>of</strong> DNA content insupraglottic epidermoid carcinoma. Otolaryngol <strong>Head</strong> <strong>Neck</strong>Surg 108:706–71024. Barrett AW, Kingsmill VJ, Speight PM (1998) The frequency<strong>of</strong> fungal infection in biopsies <strong>of</strong> oral mucosal lesions. OralDis 4:26–31


30 N. Gale · N. Zidar125. Barzan L, Talamini R (1996) Analysis <strong>of</strong> prognostic factorsfor recurrence after neck dissection. Arch Otolaryngol <strong>Head</strong><strong>Neck</strong> Surg 122:1299–130226. Batsakis JG (1999) Review article. Surgical excision margins.A pathologist’s perspective. Adv Anat Pathol 3:140–14827. Batsakis JG, Huser J (1990) Squamous carcinomas withgl<strong>and</strong>like (adenoid) features. Ann Otol Rhinol Laryngol99:87–8828. Batsakis JG, Raymond AK, Rice DH (1983) The pathology <strong>of</strong>head <strong>and</strong> neck tumors: papillomas <strong>of</strong> <strong>the</strong> upper aerodigestivetracts. <strong>Head</strong> <strong>Neck</strong> Surg 5:332–34429. Batsakis JG, Rice DH, Howard DR (1982) The pathology <strong>of</strong>head <strong>and</strong> neck tumors: spindle cell lesions (sarcomatoid carcinomas,nodular fasciitis <strong>and</strong> fibrosarcoma) <strong>of</strong> <strong>the</strong> aerodigestivetracts. <strong>Head</strong> <strong>Neck</strong> Surg 4:499–51330. Batsakis JG, Suarez P (2000) Papillary squamous carcinomas:will <strong>the</strong> real one please st<strong>and</strong> up? Adv Anat Pathol 7:2–831. Batsakis JG, Suarez P (2000) Sarcomatoid carcinomas <strong>of</strong> <strong>the</strong>upper aerodigestive tract. Adv Anat Pathol 7:282–29332. Batsakis JG, Suarez P, el-Naggar AK (1999) Proliferativeverrucous leukoplakia <strong>and</strong> its related lesions. Oral Oncol35:354–35933. Battifora H (1976) Spindle cell carcinoma: ultrastructuralevidence <strong>of</strong> squamous origin <strong>and</strong> collagen production by tumorcells. Cancer 37:2275–228234. Bauman NM, Smith RJ (1996) Recurrent respiratory papillomatosis.Pediatr Clin North Am 43:1385–140135. Becker MT, Shores CG, Yu KK, Yarbrough WG (2004) Molecularassay to detect metastatic head <strong>and</strong> neck squamouscell carcinoma. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 130:21–2736. Ber<strong>the</strong>let E, Shenouda G, Black MJ, Picariello M, Rochon L(1994) Sarcomatoid carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Am JSurg 168:455–45837. Blackburn TK, Macpherson D, Conroy B (1999) Primary adenoidsquamous cell carcinoma <strong>of</strong> <strong>the</strong> upper lip associatedwith locoregional metastasis: a case report <strong>and</strong> review <strong>of</strong> <strong>the</strong>literature. J Oral Maxill<strong>of</strong>ac Surg 57:612–61638. Blackwell KE, Calcaterra TC, Fu YS (1995) Laryngeal dysplasia:epidemiology <strong>and</strong> treatment outcome. Ann Otol RhinolLaryngol 104:596–60239. Blackwell KE, Fu YS, Calcaterra TC (1995) Laryngeal dysplasia.A clinicopathologic study. Cancer 75:457–46340. Boerman RH, Maasen EM, Joonsten J, Ka<strong>and</strong>ers HAM,Marres AM, van Overbeeke J, De Wilde P (1999) Trigeminalneuropathy secondary to perineural invasion <strong>of</strong> head <strong>and</strong>neck carcinomas. Neurology 53:213–21741. Bombi JA, Riverola A, Bordas JM, Cardesa A (1991) Adenosquamouscarcinoma <strong>of</strong> <strong>the</strong> esophagus. A case report. PatholRes Pract 187:514–51942. Boring CC, Squires TS, Tong T (1991) Cancer statistics, 1991.CA Cancer J Clin 41:19–3643. Boston M, Derkay CS (2003) Recurrent respiratory papillomatosis.Clin Pulm Med 10:10–1644. Boswell JT, Helwig EB (1965) Squamous cell carcinoma <strong>and</strong>adenoacanthoma <strong>of</strong> <strong>the</strong> stomach. A clinicopathologic study.Cancer 18:181–19245. Bouda M, Gorgoulis VG, Kastrinakis NG, Giannoudis A,Tsoli E, Danassi-Afentaki D, Foukas P, Kyroudi A, LaskarisG, Herrington CS, Kittas C (2000) “High risk” HPV typesare frequently detected in potentially malignant <strong>and</strong> malignantoral lesions, but not in normal oral mucosa. Mod Pathol13:644–65346. Bouquot JE (1994) Oral leukoplakia <strong>and</strong> erythroplakia: a review<strong>and</strong> update. Pract Periodontics Aes<strong>the</strong>t Dent 6:9–1747. Bouquot JE, Ephros H (1995) Erythroplakia: <strong>the</strong> dangerousred mucosa. Pract Periodontics Aes<strong>the</strong>t Dent 7:59–6748. Bouquot JE, Gnepp DR (1991) Laryngeal precancer: a review<strong>of</strong> <strong>the</strong> literature, commentary, <strong>and</strong> comparison with oral leukoplakia.<strong>Head</strong> <strong>Neck</strong> 13:488–49749. Bouquot JE, Gorlin RJ (1986) Leukoplakia, lichen planus,<strong>and</strong> o<strong>the</strong>r oral keratoses in 23,616 white Americans over <strong>the</strong>age <strong>of</strong> 35 years. Oral Surg Oral Med Oral Pathol 61:373–38150. Braakhuis BJM, Tabor MP, Leemans CR, Waal I van der,Snow GB, Brakenh<strong>of</strong>f RH (2002) Second primary tumours<strong>and</strong> field cancerization in oral <strong>and</strong> oropharyngeal cancer:molecular techniques provide new insights <strong>and</strong> definitions.<strong>Head</strong> <strong>Neck</strong> 24:198–20651. Brambilla E, Moro D, Veale D, Brichon PY, Stoebner P, ParamelleB, Brambilla C (1992) Basal cell (basaloid) carcinoma<strong>of</strong> <strong>the</strong> lung: a new morphologic <strong>and</strong> phenotypic entity withseparate prognostic significance. Hum Pathol 23:993–100352. Br<strong>and</strong>sma JL, Steinberg BM, Abramson AL, Winkler B (1986)Presence <strong>of</strong> human papillomavirus type 16 related sequencesin verrucous carcinoma <strong>of</strong> <strong>the</strong> larynx. Cancer Res 46:2185–218853. Brennan JA, Mao L, Hruban RH, Boyle JO, Eby YJ, KochWM, Goodman SN, Sidransky D (1995) Molecular assessment<strong>of</strong> histopathological staging in squamous-cell carcinoma<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. N Eng J Med 332:429–43554. Brito H, Vassallo J, Altemani A (2000). Detection <strong>of</strong> humanpapillomavirus in laryngeal squamous dysplasia <strong>and</strong> carcinoma.An in situ hybridization <strong>and</strong> signal amplificationstudy. Acta Otolaryngol 120:540–54455. Browson RC, Chang JC (1987) Exposure to alcohol <strong>and</strong> tobacco<strong>and</strong> <strong>the</strong> risk <strong>of</strong> laryngeal cancer. Arch Environ Health42:192–19656. Bryan RL, Bevan IS, Crocker J, Young LS (1990) Detection <strong>of</strong>HPV 6 <strong>and</strong> 11 in tumors <strong>of</strong> <strong>the</strong> upper respiratory tract using<strong>the</strong> polymerase chain reaction. Clin Otolaryngol 15:177–18057. Bryne M (1998) Is <strong>the</strong> invasive front <strong>of</strong> an oral carcinoma <strong>the</strong>most important area for prognostication? Oral Dis 4:70–7758. Bryne M, Jenssen N, Boysen M (1995) Histologic gradingin <strong>the</strong> deep invasive front <strong>of</strong> T1 <strong>and</strong> T2 glottic squamouscell carcinomas has high prognostic value. Virchows Arch427:277–28159. Calhoun KH, Fulmer P, Weiss R, Hokanson JA (1994) Distantmetastases from head <strong>and</strong> neck squamous cell carcinomas.Laryngoscope 104:1199–120560. Califano J, van der Riet P, Westra W, Nawroz H, ClaymanG, Piantadosi S, Corio R, Lee D, Greenberg B, Koch W, SidranskyD (1996) Genetic progression model for head <strong>and</strong>neck cancer: implications for field cancerization. Cancer Res56:2488–249261. Carlos R, Sedano HO (1994) Multifocal papilloma virus epi<strong>the</strong>lialhyperplasia. Oral Surg Oral Med Oral Pathol 77:631–63562. Carter RL, Barr LC, O’Brien CJ, Soo KC, Shaw HK (1985)Transcapsular spread <strong>of</strong> metastatic squamous cell carcinomafrom cervical lymph nodes. Am J Surg 150:495–49963. Cazorla M, Hern<strong>and</strong>ez L, Nadal A, Balbin M, Lopez JM, VizosoF, Fern<strong>and</strong>ez PL, Iwata K, Cardesa A, Lopez-Otin C,Campo E (1998) Collagenase-3 expression is associated withadvanced local invasion in human squamous cell carcinomas<strong>of</strong> <strong>the</strong> larynx. J Pathol 186:144–15064. Chen YK, Hsue SS, Lin LM (2003) Immunohistochemicaldemonstration <strong>of</strong> p63 in DMBA-induced hamster buccalpouch squamous cell carcinogenesis Oral Dis 9:235–24065. Cho KJ, Jang JJ, Lee SS, Zo JI (2000) Basaloid squamous carcinoma<strong>of</strong> <strong>the</strong> oesophagus: a distinct neoplasm with multipotentialdifferentiation. Histopathology 36:331–34066. Choi HR, Sturgis EM, Rosenthal DI, Luna MA, Batsakis JG,El-Naggar AK (2003) Sarcomatoid carcinoma <strong>of</strong> <strong>the</strong> head<strong>and</strong> neck. Molecular evidence for evolution <strong>and</strong> progressionfrom conventional squamous cell carcinoma. Am J SurgPathol 27:1216–122067. Chow TL, Chow TK, Lui YH, Sze WM, Yuen NWF, KwokSPY (2002) Lymphoepi<strong>the</strong>lioma-like carcinoma <strong>of</strong> oral cavity:report <strong>of</strong> three cases <strong>and</strong> literature review. Int J Oral Maxill<strong>of</strong>acSurg 31:212–21868. Cihak RW, Kawashima T, Steer A (1972) Adenoacanthoma(adenosquamous carcinoma) <strong>of</strong> <strong>the</strong> pancreas. Cancer29:1133–114069. Coletta RD, Cotrim P, Almeida OP, Alves VAF, WakamatsuA, Vargas PA (2002) Basaloid squamous carcinoma <strong>of</strong> oralcavity: a histologic <strong>and</strong> immunohistochemical study. OralOncol 38:723–729


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 3170. Cook JR, Hill DA, Humphrey PA, Pfeifer JD, El-M<strong>of</strong>ty SK(2000) Squamous cell carcinoma arising in recurrent respiratorypapillomatosis with pulmonary involvement: emergingcommon pattern <strong>of</strong> clinical features <strong>and</strong> human papillomavirusserotype association. Mod Pathol 13:914–91871. Cooper JR, Hellquist H, Michaels L (1992) Image analysis in<strong>the</strong> discrimination <strong>of</strong> verrucous carcinoma <strong>and</strong> squamouspapilloma. J Pathol 166:383–38772. Coppola D, Catalano E, Tang CK, Elfenbein IB, Harwick R,Mohr R (1993) Basaloid squamous cell carcinoma <strong>of</strong> floor <strong>of</strong>mouth. Cancer 72:2299–230573. Cör A, Gale N, Kambič V (1997) Quantitative pathology <strong>of</strong>laryngeal epi<strong>the</strong>lial hyperplastic lesions. Acta Otolaryngol527:57–6174. Crissman JD, Gluckman JL, Whitely J, Quernelle D (1980)Squamous cell carcinoma <strong>of</strong> <strong>the</strong> floor <strong>of</strong> <strong>the</strong> mouth. <strong>Head</strong><strong>Neck</strong> Surg 3:2–775. Crissman JD, Kessis T, Shah KV, Fu YS, Stoler MH, Zarbo RJ,Weiss MA (1988) Squamous papillary neoplasia <strong>of</strong> <strong>the</strong> adultupper aerodigestive tract. Hum Pathol 19:1387–139676. Crissman JD, Liu MY, Gluckman J, Cummings G (1984).Prognostic value <strong>of</strong> histopathologic parameters in squamouscell carcinoma <strong>of</strong> <strong>the</strong> oropharynx. Cancer 54:2995–300177. Crissman JD, Sakr WA (2001) Squamous neoplasia <strong>of</strong> <strong>the</strong> upperaerodigestive tract. In: Pilch BZ (ed) <strong>Head</strong> <strong>and</strong> neck surgicalpathology. Lippincott Williams & Wilkins, Philadelphia,pp 34–5278. Crissman JD, Visscher DW, Sakr W (1993) Premalignant lesions<strong>of</strong> <strong>the</strong> upper aerodigestive tract: pathologic classification.J Cell Biochem 17:49–5679. Crissman JD, Zarbo RJ (1989) Dysplasia, in situ carcinoma,<strong>and</strong> progression to invasive squamous cell carcinoma <strong>of</strong> <strong>the</strong>upper aerodigestive tract. Am J Surg Pathol 13:5–1680. Crissman JD, Zarbo RJ, Drozdowicz S, Jacobs J, Ahmad K,Weaver A (1988) Carcinoma in situ <strong>and</strong> microinvasive squamouscarcinoma <strong>of</strong> <strong>the</strong> laryngeal glottis. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 114:299–30781. Cromer A, Carles A, Millon R, Ganguli G, Chalmel F, LemaireF, Young J, Dembélé D, Thibault C, Muller D, Poch O,Abecassis J, Wasylyk B (2004) Identification <strong>of</strong> genes associatedwith tumorigenesis <strong>and</strong> metastatic potential <strong>of</strong> hypopharyngealcancer by microarray analysis. Oncogene23:2484–249882. Cruz IB, Snijders PJF, Meijer CJ, Braakhuis BJ, Snow GB,Walboomers JM, van der Waal I (1998) P53 expression above<strong>the</strong> basal cell layer in oral mucosa is an early event <strong>of</strong> malignanttransformation <strong>and</strong> has predictive value for developingoral squamous cell carcinoma. J Pathol 184:360–36883. Damiani JM, Damiani KK, Hauck K, Hyams VJ (1981) Mucoepidermoid-adenosquamouscarcinoma <strong>of</strong> <strong>the</strong> larynx <strong>and</strong>hypopharynx: a report <strong>of</strong> 21 cases <strong>and</strong> a review <strong>of</strong> <strong>the</strong> literature.Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 89:235–24384. De Bree R, Mehta DM, Snow GB, Quak JJ (2001) Intracranialmetastases in patients with squamous cell carcinoma <strong>of</strong> <strong>the</strong>head <strong>and</strong> neck. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 124:217–22185. De Carvalho MB (1998) Quantitive analysis <strong>of</strong> <strong>the</strong> extent <strong>of</strong>extracapsular invasion <strong>and</strong> its prognostic significance: a prospectivestudy <strong>of</strong> 170 cases <strong>of</strong> carcinoma <strong>of</strong> <strong>the</strong> larynx <strong>and</strong>hypopharynx. <strong>Head</strong> <strong>Neck</strong> 20:16–2186. De Stefani E, Oreggia F, Rivero S, Fierro L (1992) H<strong>and</strong>-rolledcigarette smoking <strong>and</strong> risk <strong>of</strong> cancer <strong>of</strong> <strong>the</strong> mouth, pharynx<strong>and</strong> larynx. Cancer 70:679–68287. Derkay CS (2001) Recurrent respiratory papillomatosis. Laryngoscope111:57–6988. Derkay CS, Darrow DH (2000) Recurrent respiratory papillomatosis<strong>of</strong> <strong>the</strong> larynx: current diagnosis <strong>and</strong> treatment.Otolaryngol Clin North Am 33:1127–114289. Derkay CS, Rimell FL, Thompson JW (1998) Recurrent respiratorypapillomatosis. <strong>Head</strong> <strong>Neck</strong> 20:418–42490. Dougherty BG, Evans HL (1985) Carcinoma <strong>of</strong> <strong>the</strong> anal canal:a study <strong>of</strong> 79 cases. Am J Clin Pathol 83:159–16491. Doyle DJ, Gianoli GJ, Espinola T, Miller RH (1994) Recurrentrespiratory papillomatosis: juvenile versus adult forms.Laryngoscope 104:523–52792. Doyle DJ, Henderson LA, LeJeune FE Jr, Miller RH (1994)Changes in human papillomavirus typing <strong>of</strong> recurrent respiratorypapillomatosis progressing to malignant neoplasm.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 120:1273–127693. Dubey P, Ha CS, Ang KK, El-Naggar AK, Knapp C, ByersRM, Morrison WH (1998) Nonnasopharyngeal lymphoepi<strong>the</strong>lioma<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Cancer 82:1556–156294. Dvorak HF (1986) Tumors: wounds that do not heal. N EnglJ Med 315:1650–165995. Edstrom S, Johansson SL, Lindstrom J, S<strong>and</strong>in I (1987) Verrucoussquamous cell carcinoma <strong>of</strong> <strong>the</strong> larynx: evidence forincreased metastatic potential after irradiation. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 97:381–38496. Ellis GL, Langloss JM, Heffner DK, Hyams VJ (1987) Spindle-cellcarcinoma <strong>of</strong> <strong>the</strong> aerodigestive tract. An immunohistochemicalanalysis <strong>of</strong> 21 cases. Am J Surg Pathol 11:335–34297. El-M<strong>of</strong>ty SK, Lu DW (2003) Prevalence <strong>of</strong> human papillomavirustype 16 DNA in squamous cell carcinoma <strong>of</strong> <strong>the</strong> palatinetonsil, <strong>and</strong> not <strong>the</strong> oral cavity, in young patients: a distinctclinicopathologic <strong>and</strong> molecular disease entity. Am JSurg Pathol 27:1463–147098. El-Naggar AK, Dinh M, Tucker S, Luna MA, Goepfert H, HsuP, Batsakis JG (1996) Genotypic analysis <strong>of</strong> primary head <strong>and</strong>neck squamous carcinoma by combined fluorescence in situhybridization <strong>and</strong> DNA flow cytometry. Am J Clin Pathol105:102–10899. Ereño C, Lopez JI, Sanchez JM, Bilbao FJ (2001) Papillarysquamous cell carcinoma <strong>of</strong> <strong>the</strong> larynx. J Laryngol Otol115:164–166100. Eversole LR, Laipis PJ, Merrell P, Choi E (1987) Demonstration<strong>of</strong> human papillomavirus DNA in oral condyloma acuminatum.J Oral Pathol 16:266–272101. Fagan JJ, Collins B, Barnes L, D’Amico F, Myers EN, JohnsonJT (1998) Perineural invasion in squamous cell carcinoma<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg124:637–640102. Faridoni-Laurens L, Bosq J, Janot F, Vayssade M, Le BihanML, Kaghad M, Caput D, Benard J, Ahomadegbe JC (2001)P73 expression in basal layers <strong>of</strong> head <strong>and</strong> neck squamous epi<strong>the</strong>lium:a role in differentiation <strong>and</strong> carcinogenesis in concertwith p53 <strong>and</strong> p63? Oncogene 20:5302–5312103. Ferlito A, Altavilla G, Rinaldo A, Doglioni C (1997) Basaloidsquamous cell carcinoma <strong>of</strong> <strong>the</strong> larynx <strong>and</strong> hypopharynx.Ann Otol Rhinol Laryngol 106:1024–1035104. Ferlito A, Devaney KO, Devaney SL, Rinaldo A, Carbone A(1999) Clinicopathological consultation. Micrometastases:have <strong>the</strong>y an impact on prognosis? Ann Otol Rhinol Laryngol108:1185–1189105. Ferlito A, Devaney KO, Rinaldo A, Milroy CM, Carbone A(1996) Clinicopathological consultation. Mucosal adenoidsquamous cell carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Ann OtolRhinol Laryngol 105:409–413106. Ferlito A, Partridge M, Brennan J, Hamakawa H (2001)Lymph node micrometastasis in <strong>the</strong> head <strong>and</strong> neck cancer: areview. Acta Otolaryngol 121:660–665107. Ferlito A, Recher G (1980) Ackerman’s tumor (verrucous carcinoma)<strong>of</strong> <strong>the</strong> larynx: a clinicopathologic study <strong>of</strong> 77 cases.Cancer 46:1617–1630108. Ferlito A, Rinaldo A, Devaney KO, Devaney SL, Milroy CM(1998) Impact <strong>of</strong> phenotype on treatment <strong>and</strong> prognosis <strong>of</strong>laryngeal malignancies. J Laryngol Otol 112:710–714109. Ferlito A, Rinaldo A, Devaney KO, MacLennan K, Myers JN,Petruzzelli GJ, Shaha AR, Genden EM, Johnson JT, de CarvalhoMB, Myers EN (2002) Prognostic significance <strong>of</strong> microscopic<strong>and</strong> macroscopic extracapsular spread from metastatictumor in <strong>the</strong> cervical lymph nodes. Oral Oncol 38:747–751110. Ferlito A, Rinaldo A, Devaney KO, Putzi MT (1999) Papillarysquamous cell carcinoma versus verrucous squamouscell carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Ann Otol Rhinol Laryngol108:318–322111. Ferlito A, Rinaldo A, Robbins KT, Leemans CR, Shah JP, ShahaAR, Andersen PE, Kowalski LP, Pellitteri PK, ClaymanGL, Rogers SN, Medina JE, Byers RM (2003) Changing con-


32 N. Gale · N. Zidar1cepts in <strong>the</strong> surgical management <strong>of</strong> <strong>the</strong> cervical node metastasis.Oral Oncol 39:429–435112. Ferlito A, Shaha AR, Silver CE, Rinaldo A, Mondin V (2001)Incidence <strong>and</strong> sites <strong>of</strong> distant metastases from head <strong>and</strong> neckcancer. ORL J Otorhinolaryngol Relat Spec 63:202–207113. Ferlito A, Weiss CM, Rinaldo A, Carbone A, Devaney KO,MacMillan C, Barnes L (1997) Clinicopathologic consultation.Lymphoepi<strong>the</strong>lial carcinoma <strong>of</strong> <strong>the</strong> larynx, hypopharynx<strong>and</strong> trachea. Ann Otol Rhinol Laryngol 106:437–444114. Fettig A, Pogrel MA, Silverman S Jr, Bramanti TE, Da CostaM, Regezi JA (2000) Proliferative verrucous leukoplakia <strong>of</strong><strong>the</strong> gingiva. Oral Surg Oral Med Oral Pathol Oral Radiol Endod90:723–730115. Fiorella R, Di Nicola V, Resta L (1997) Epidemiological <strong>and</strong>clinical relief on hyperplastic lesions <strong>of</strong> <strong>the</strong> larynx. Acta OtolaryngolSuppl 527:77–81116. Fliss DM, Noble-Topham SE, McLachlin CM, Freeman JL,Noyek AM, van Nostr<strong>and</strong> AWP, Hartwick WJ (1994) Laryngealverrucous carcinoma: a clinicopathologic study <strong>and</strong> detection<strong>of</strong> human papillomavirus using polymerase chain reaction.Laryngoscope 104:146–152117. Forastiere A, Koch W, Trotti A, Sidransky D (2001) <strong>Head</strong> <strong>and</strong>neck cancer. N Engl J Med 345:1890–1900118. Fouret P, Dabit D, Sibony M, Alili D, Commo F, Saint-GuilyJL, Callard P (1995) Expression <strong>of</strong> p53 protein related to <strong>the</strong>presence <strong>of</strong> human papillomavirus infection in precancer lesions<strong>of</strong> <strong>the</strong> larynx. Am J Pathol 146:599–604119. Fracchiolla NS, Pignataro L, Capaccio P, Trecca D, BoletiniA, Ottaviani A, Polli E, Maiolo AT, Neri A (1995) Multiplegenetic lesions in laryngeal squamous cell carcinomas. Cancer75:1292–1301120. Frank DK, Cheron F, Cho H, DiConstanzo D, Sclafani AP(1995) Nonnasopharyngeal lymphoepi<strong>the</strong>liomas (undifferentiatedcarcinomas) <strong>of</strong> <strong>the</strong> upper aerodigestive tract. AnnOtol Rhinol Laryngol 104:305–310121. Freier K, Joos S, Flechtenmacher C, Devens F, Benner A,Bosch FX, Lichter P, H<strong>of</strong>ele C (2003) Tissue microarray analysisreveals site-specific prevalence <strong>of</strong> oncogene amplificationsin head <strong>and</strong> neck squamous cell carcinoma. Cancer Res63:1179–1182122. Friedmann I, Ferlito A (1993) Precursors <strong>of</strong> squamous cellcarcinoma. In: Ferlito’s neoplasms <strong>of</strong> <strong>the</strong> larynx. ChurchillLivingstone, Edinburgh, pp 97–111123. Frierson HF, Bellafiore FJ, Gaffey MJ, McCary WS, Innes DJ,Williams ME (1994) Cytokeratin in anaplastic cell large celllymphoma. Mod Pathol 7:317–321124. Fujino K, Ito J, Kanaji M, Shiomi Y, Saiga T (1995) Adenosquamouscarcinoma <strong>of</strong> <strong>the</strong> larynx. Am J Otolaryngol16:115–118125. Gale N, Kambič V, Michaels L, Cardesa A, Hellquist H, ZidarN, Poljak M (2000) The Ljubljana classification: a practicalstrategy for <strong>the</strong> diagnosis <strong>of</strong> laryngeal precancerous lesions.Adv Anat Pathol 7:240–251126. Gale N, Poljak M, Kambič V, Ferluga D, Fischinger J (1994)Laryngeal papillomatosis: molecular, histopathological, <strong>and</strong>clinical evaluation. Virchows Arch 425:291–295127. Gale N, Zidar N, Kambič V, Poljak M, Cör A (1997) Epidermalgrowth factor receptor, c-erbB-2 <strong>and</strong> p53 overexpressionsin epi<strong>the</strong>lial hyperplastic lesions <strong>of</strong> <strong>the</strong> larynx. ActaOtolaryngol 527:105–110128. Gallo O, Bianchi S, Giannini A, Boccuzzi S, Calzolari A,Fini-Storchi O (1994) Lack <strong>of</strong> detection <strong>of</strong> human papillomavirus(HPV) in transformed laryngeal keratosis by in situ hybridization(ISH) technique. Acta Otolaryngol 114:213–217129. Gallo O, Franchi A, Chiarelli I, Porfirio B, Gr<strong>and</strong>e A, SimonettiL, Bocciolini C, Fini-Storchi O (1997) Potential biomarkersin predicting progression <strong>of</strong> epi<strong>the</strong>lial hyperplasticlesions <strong>of</strong> <strong>the</strong> larynx. Acta Otolaryngol Suppl 527:30–38130. Genden EM, Ferlito A, Bradley PJ, Rinaldo A, Scully C (2003)<strong>Neck</strong> disease <strong>and</strong> distant metastases. Oral Oncol 39:207–212131. Gerughty RM, Hennigar GR, Brown FM (1968) Adenosquamouscarcinoma <strong>of</strong> <strong>the</strong> nasal, oral <strong>and</strong> laryngeal cavities: aclinicopathologic survey <strong>of</strong> ten cases. Cancer 22:1140–1155132. Gluckmann A, Cherry CP (1956) Incidence, histology, <strong>and</strong>response to radiation <strong>of</strong> mixed carcinomas (adenoacanthomas)<strong>of</strong> <strong>the</strong> uterine cervix. Cancer 9:971–979133. Go C, Schwartz MR, Donovan DT (2003) Molecular transformation<strong>of</strong> recurrent respiratory papillomatosis: viraltyping <strong>and</strong> p53 overexpression. Ann Otol Rhinol Laryngol112:298–302134. Goes FCGD, Oliviera DT, Dorta RG, Nishimoto IN, L<strong>and</strong>manG, Kowalski LP (2004) Prognoses <strong>of</strong> oral basaloid squamouscell carcinoma <strong>and</strong> squamous cell carcinoma – a comparison.Arch Otolaryngol 130:83–86135. Goldman RL, Klein HZ, Sung M (1977) Adenoid squamouscell carcinoma <strong>of</strong> <strong>the</strong> oral cavity: report <strong>of</strong> <strong>the</strong> first case arisingin <strong>the</strong> tongue. Arch Otolaryngol 103:496–498136. Gorgoulis V, Rassidakis G, Karameris A, GiatromanolakiA, Barbatis C, Kittas C (1994) Expression <strong>of</strong> p53 protein inlaryngeal squamous cell carcinoma <strong>and</strong> dysplasia: possiblecorrelation with human papillomavirus infection <strong>and</strong> clinicopathologicalfindings. Virchows Arch 425:481–489137. Gorgoulis VG, Zacharatos P, Kotsinas A, Kyroudi A, RassidakisAN, Ikonomopoulos JA, Barbatis C, Herrington CS,Kittas C (1999) Human papilloma virus (HPV) is possiblyinvolved in laryngeal but not in lung carcinogenesis. HumPathol 30:274–283138. Grasl MCH, Neuwirth-Riedl K, Vutuc C, Horak F, VorbeckF, Banyai M (1990) Risk <strong>of</strong> vocal cord dysplasia in relationto smoking, alcohol intake <strong>and</strong> occupation. Eur J Epidemiol6:45–48139. Gray MH, Rosenberg AE, Dickersin GR, Bhan AK (1990) Cytokeratinexpression in epi<strong>the</strong>loid vascular neoplasms. HumPathol 21:212–217140. Grayson W, Cooper K (2002) A reappraisal <strong>of</strong> “basaloid carcinoma”<strong>of</strong> <strong>the</strong> cervix, <strong>and</strong> <strong>the</strong> differential diagnosis <strong>of</strong> basaloidcervical neoplasms. Adv Anat Pathol 5:290–300141. Green GE, Bauman NM, Smith RJ (2000) Pathogenesis <strong>and</strong>treatment <strong>of</strong> juvenile onset recurrent respiratory papillomatosis.Otolaryngol Clin North Am 33:187–207142. Green TL, Eversole LR, Leider AS (1986) Oral <strong>and</strong> labial verrucavulgaris: clinical, histologic <strong>and</strong> immunohistochemicalevaluation. Oral Surg Oral Med Oral Pathol 62:410–416143. Guillou L, Sahli R, Chaubert P, Monnier P, Cuttat JF, CostaJ (1991) Squamous cell carcinoma <strong>of</strong> <strong>the</strong> lung in a nonsmoking,nonirradiated patient with juvenile laryngotracheal papillomatosis.Evidence <strong>of</strong> human papillomavirus-11 DNA inboth carcinoma <strong>and</strong> papillomas. Am J Surg Pathol 15:891–898144. Ha PK, Califano JA (2002) The molecular biology <strong>of</strong> laryngealcancer. Otolaryngol Clin North Am 35:993–1012145. Hagen P, Lyons GD, Haindel C (1993) Verrucous carcinoma<strong>of</strong> <strong>the</strong> larynx: role <strong>of</strong> human papillomavirus, radiation, <strong>and</strong>surgery. Laryngoscope 103:253–257146. Hamakawa H, Fukizumi M, Bao Y, Sumida T, Onishi A,Tanioka H, Sato H, Yumoto E (1999) Genetic diagnosis <strong>of</strong>micrometastasis based on SCC antigen mRNA in cervicallymph nodes <strong>of</strong> head <strong>and</strong> neck cancer. Clin Exp Metastasis17:593–599147. Hamakawa H, Takemura K, Sumida T, Kayahara H, TaniokaH, Sogawa K (2000) Histological study on pN upgrading <strong>of</strong>oral cancer. Virchows Arch 437:116–121148. Hannen EJM, Macville MVE, Wienk SM, Slootweg PJ, ManniJJ, Hanselaar AGJM, de Wilde PCM (2004) Different chromosomalimbalances in metastasized <strong>and</strong> nonmetastasizedtongue carcinomas identified by comparative genomic hybridization.Oral Oncol 40:364–371149. Haughey BH, Gates GA, Arkfen CL, Harvey J (1992) Metaanalysis<strong>of</strong> second tumours in head <strong>and</strong> neck cancer: <strong>the</strong> casefor an endoscopic screening protocol. Ann Otol Rhinol Laryngol101:105–112150. Hellquist H, Cardesa A, Gale N, Kambič V, Michaels L (1999)Criteria for grading in <strong>the</strong> Ljubljana classification <strong>of</strong> epi<strong>the</strong>lialhyperplastic laryngeal lesions. A study by members <strong>of</strong><strong>the</strong> Working group on Epi<strong>the</strong>lial Hyperplastic Laryngeal Lesions<strong>of</strong> <strong>the</strong> European Society <strong>of</strong> <strong>Pathology</strong>. Histopathology34:226–233


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 33151. Hellquist H, Ol<strong>of</strong>sson J (1989) Spindle cell carcinoma <strong>of</strong> <strong>the</strong>larynx. APMIS 97:1103–1113152. Hellquist HB, Ol<strong>of</strong>sson J (1988) Expression <strong>of</strong> low molecularweight cytokeratin proteins in laryngeal dysplasia. AP-MIS 96:971–978153. Herrmann K, Niedobitek G (2003) Epstein-Barr virus-associatedcarcinomas; facts <strong>and</strong> fiction. J Pathol 199:140–145154. Hewan-Lowe K, Dardick I (1995) Ultrastructural distinction<strong>of</strong> basaloid-squamous carcinoma <strong>and</strong> adenoid cystic carcinoma.Ultrastruct Pathol 19:371–381155. Hirabayashi H, Koshii K, Uno K, Ohgaki H, Nakasone Y, FujisawaT, Syouno N, Hinohara T, Hirabayashi K (1991) Extracapsularspread <strong>of</strong> squamous cell carcinoma in neck lymphnodes: prognostic factor <strong>of</strong> laryngeal cancer. Laryngoscope101:501–506156. Hirai T, Hayashi K, Takumida M, Ueda T, Hirakawa K, YajinK (2003) Reduced expression <strong>of</strong> p27 is correlated with progressionin precancerous lesions <strong>of</strong> <strong>the</strong> larynx. Auris NasusLarynx 30:163–168157. H<strong>of</strong>fman GR, Hayter JP (2002) Widespread subcutaneousdistant metastases from a head <strong>and</strong> neck squamous cell carcinoma.J Oral Maxill<strong>of</strong>ac Surg 60:954–958158. Holmgren L, O’Reilly S, Folkman J (1995) Dormancy <strong>of</strong> micrometastasis:balanced proliferation <strong>and</strong> apoptosis in <strong>the</strong>presence <strong>of</strong> angiogenesis. Nat Med 1:149–153159. Ide F, Shimoyama T, Horie N, Kusama K (2002) Basaloidsquamous cell carcinoma <strong>of</strong> <strong>the</strong> oral mucosa: new case <strong>and</strong>review <strong>of</strong> 45 cases in <strong>the</strong> literature. Oral Oncol 38:120–124160. Iezzoni JC, Gaffey MJ, Weiss LM (1995) The role <strong>of</strong> Epstein-Barr virus in lymphoepi<strong>the</strong>lioma-like carcinomas. Am J ClinPathol 103:308–315161. Ishiyama A, Eversole LR, Ross DA, Raz Y, Kerner MM, FuYS, Blackwell KE, Feneberg R, Bell TS, Calcaterra TC (1994)Papillary squamous neoplasms <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Laryngoscope104:1446–1452162. Izawa H, Yonemitsu N, Shin T, Sugihara H (1999) Histopathologicalanalysis <strong>of</strong> apoptosis, <strong>and</strong> expression <strong>of</strong> p53, bcl-2,bax, <strong>and</strong> Ki-67 in laryngeal squamous cell carcinomas <strong>and</strong>dysplasia. Auris Nasus Larynx 26:317–330163. Izbicki JR, Pantel K, Hosch SB (2002) Micrometastasis insolid epi<strong>the</strong>lial tumors: impact on surgical oncology. Surgery131:1–5164. Izumi K, Nakajima T, Maeda T, Cheng J, Saku T (1998) Adenosquamouscarcinoma <strong>of</strong> <strong>the</strong> tongue – report <strong>of</strong> a case withhistochemical, immunohistochemical, <strong>and</strong> ultrastructuralstudy <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Oral Surg Oral Med OralPathol Oral Radiol Endod 85:178–184165. Jaber MA, Porter SR, Gilthorpe MS, Bedi R, Scully C (1999)Risk factors for oral epi<strong>the</strong>lial dysplasia – <strong>the</strong> role <strong>of</strong> smoking<strong>and</strong> alcohol. Oral Oncol 35:151–156166. Janot F, Klijanienko J, Russo, Mamet JP, de Braud F, El-NaggarAK, Pignon JP, Luboinski B, Cvitkovic E (1996) Prognosticvalue <strong>of</strong> clinicopathologic parameters in head <strong>and</strong> necksquamous carcinoma: a prospective analysis. Br J Cancer73:531–538167. Jares P, Fernández PL, Campo E, Nadal A, Bosch F, Aiza G,Nayach I, Traserra J, Cardesa A (1994) PRAD-1/Cyclin D1gene amplification correlates with messenger RNA overexpression<strong>and</strong> tumor progression in human laryngeal carcinomas.Cancer Res 54:4813–4817168. Jares P, Fernández PL, Nadal A, Cazorla M, Hernández L,Pinyol M, Hernández S, Traserra J, Cardesa A, Campo E(1997) p16MTS1/CDK4I mutations <strong>and</strong> concomitant loss <strong>of</strong>heterozygosity at 9p21-23 are frequent events in squamouscell carcinoma <strong>of</strong> <strong>the</strong> larynx. Oncogene 15:1445–1453169. Jares P, Nadal A, Fern<strong>and</strong>ez PL, Pinyol M, Hern<strong>and</strong>ez L, CazorlaM, Hern<strong>and</strong>ez S, Bea S, Cardesa A, Campo E (1999)Disregulation <strong>of</strong> p16MTS1/CDK4I protein <strong>and</strong> mRNA expressionis associated with gene alterations in squamous-cellcarcinoma <strong>of</strong> <strong>the</strong> larynx. Int J Cancer 81:705–711170. Johnson NW, Ranasinghe AW, Warnakulasuriya KA (1993)Potentially malignant lesions <strong>and</strong> conditions <strong>of</strong> <strong>the</strong> mouth<strong>and</strong> oropharynx: natural history-cellular <strong>and</strong> molecularmarkers <strong>of</strong> risk. Eur J Cancer Prev 2:31–51171. Johnson NW, Warnakulasuriy S, Tavassoli M (1996) Hereditary<strong>and</strong> environmental risk factors; clinical <strong>and</strong> laboratoryrisk matters for head <strong>and</strong> neck, especially oral, cancer <strong>and</strong>precancer. Eur J Cancer Prev 5:5–17172. Johnson TL, Plieth DA, Crissman JD, Sarkar FH (1991) HPVdetection by polymerase chain reaction (PCR) in verrucouslesions <strong>of</strong> <strong>the</strong> upper aerodigestive tract. Mod Pathol 4:461–463173. Jones AC, Freedman PD, Kerpel SM (1993) Oral adenoidsquamous cell carcinoma: a report <strong>of</strong> three cases <strong>and</strong> review<strong>of</strong> <strong>the</strong> literature. J Oral Maxill<strong>of</strong>ac Surg 51:676–681174. Jones AS, Morar P, Phillips DE, Field JK, Husb<strong>and</strong> D, HelliwellTR (1994) Second primary tumours in patients withhead <strong>and</strong> neck cancer. Cancer 95:1343–1352175. Jose J, Coatesworth AP, MacLennan K (2003) Cervical metastasesin upper aerodigestive tract squamous cell carcinoma:histopathologic analysis <strong>and</strong> reporting. <strong>Head</strong> <strong>Neck</strong>25:194–197176. Jose J, Moor JW, Coatesworth AP, Johnston C, MacLennanK (2004) S<strong>of</strong>t tissue deposits in neck dissections <strong>of</strong> patientswith head <strong>and</strong> neck squamous cell carcinoma. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 130:157–160177. Jyotirmayi R, Sankaranarayanan R, Varghese C, Jacob R,Krishnan Nair M (1997) Radio<strong>the</strong>rapy in <strong>the</strong> treatment <strong>of</strong>verrucous carcinoma <strong>of</strong> <strong>the</strong> oral cavity. Oral Oncol 33:124–128178. Kambič V (1978) Difficulties in management <strong>of</strong> vocal cordprecancerous lesions. J Laryngol Otol 92:305–315179. Kambič V (1997) Epi<strong>the</strong>lial hyperplastic lesions – a challengingtopic in laryngology. Acta Otolaryngol 527:7–11180. Kambič V, Gale N (1986) Significance <strong>of</strong> keratosis <strong>and</strong> dyskeratosisfor classifying hyperplastic aberrations <strong>of</strong> laryngealmucosa. Am J Otolaryngol 7:323–333181. Kambič V, Gale N (1995) Epi<strong>the</strong>lial hyperplastic lesions <strong>of</strong><strong>the</strong> larynx. Elsevier, Amsterdam, pp 1–265182. Kambič V, Gale N, Ferluga D (1992) Laryngeal hyperplasticlesions, follow-up study <strong>and</strong> application <strong>of</strong> lectins <strong>and</strong> anticytokeratinsfor <strong>the</strong>ir evaluation. Pathol Res Pract 188:1067–1077183. Kambič V, Lenart I (1971) Our classification <strong>of</strong> hyperplasia <strong>of</strong><strong>the</strong> laryngeal epi<strong>the</strong>lium from <strong>the</strong> prognostic point <strong>of</strong> view. JFr Otorhinolaryngol Audiophonol Chir Maxill<strong>of</strong>ac 20:1145–1150184. Kambič V, Radšel Z, Gale N (1989) Alterations in <strong>the</strong> laryngealmucosa after exposure to asbestos. Br J Ind Med 46:717–723185. Kambič V, Radšel Z, Preželj J, Žargi M (1984) The role <strong>of</strong>testosterone in laryngeal carcinogenesis. Am J Otolaryngol5:344–349186. Kansky AA, Poljak M, Seme K, Kocjan BJ, Gale N, Luzar B,Golouh R (2003) Human papillomavirus DNA in oral squamouscell carcinomas <strong>and</strong> normal oral mucosa. Acta Virol47:11–16187. Kashima H, Mounts P, Leventhal B, Hruban RH (1993) Sites<strong>of</strong> predilection in recurrent respiratory papillomatosis. AnnOtol Rhinol Laryngol 102:580–583188. Kashima HK, Mounts P, Shah K (1996) Recurrent respiratorypapillomatosis. Obstet Gynecol Clin North Am 23:699–706189. Kashima HK, Shah F, Lyles A, Glackin R, Muhammad N,Turner L, Van Z<strong>and</strong>t S, Whitt S, Shah K (1992) A comparison<strong>of</strong> risk factors in juvenile-onset <strong>and</strong> adult-onset recurrent respiratorypapillomatosis. Laryngoscope 102:9–13190. Kasperbauer JL, O’Halloran GL, Espy MJ, Smith TF, LewisJE (1993) Polymerase chain reaction (PCR) identification <strong>of</strong>human papillomavirus (HPV) DNA in verrucous carcinoma<strong>of</strong> <strong>the</strong> larynx. Laryngoscope 103:416–420191. Kaugars GE, Silverman S Jr, Lovas JG, Thompson JS, Br<strong>and</strong>tRB, Singh VN (1996) Use <strong>of</strong> antioxidant supplements in <strong>the</strong>treatment <strong>of</strong> human oral leukoplakia. Oral Surg Oral MedOral Pathol Oral Radiol Endod 81:5–14192. Keelawat S, Liu CZ, Roehm PC, Barnes L (2002) Adenosquamouscarcinoma <strong>of</strong> <strong>the</strong> upper aerodigestive tract: a clinicopathologicstudy <strong>of</strong> 12 cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. AmJ Otolaryngol 23:160–168


34 N. Gale · N. Zidar1193. Kell MR, Winter DC, O’Sullivan GC, Shanaha F, RedmondHP (2000) Biological behaviour <strong>and</strong> clinical implications <strong>of</strong>micrometastasis. Br J Surg 87:1629–1639194. Kleinsasser O (1988) Tumors <strong>of</strong> <strong>the</strong> larynx <strong>and</strong> hypopharynx.Thieme, Stuttgart, pp 61–123195. Klijanienko J, El-Naggar A, Ponzio-Prion A, Mar<strong>and</strong>as P,Micheau C, Caillaud JM (1993) Basaloid squamous carcinoma<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Immunohistochemical comparisonwith adenoid cystic carcinoma <strong>and</strong> squamous cell carcinoma.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 119:887–890196. Koch BB, Trask DK, H<strong>of</strong>fman HT, Karnell LH, Robinson RA,Zhen W, Menck HR (2001) National survey <strong>of</strong> head <strong>and</strong> neckverrucous carcinoma: pattern <strong>of</strong> presentation, care, <strong>and</strong> outcome.Cancer 92:110–120197. Kolbusz RV, Goldberg LH (1994) Verrucous carcinoma <strong>of</strong> <strong>the</strong>oral cavity. Int J Dermatol 33:618–622198. Kotwall C, Sako K, Razack MS, Rao U, Bakamjian V, SheddDP (1987) Metastatic patterns in squamous cell cancer <strong>of</strong> <strong>the</strong>head <strong>and</strong> neck. Am J Surg 154:439–442199. Krecicki T, Jelen M, Zalesska-Krecicka M, Szkudlarek T, SzajowskiK (1999) Immunohistochemically stained markers(p53, PCNA, bcl-2) in dysplastic lesions <strong>of</strong> <strong>the</strong> larynx. CancerLett 143:23–28200. Küffer R, Lombardi T (2002) Premalignant lesions <strong>of</strong> <strong>the</strong> oralmucosa. A discussion about <strong>the</strong> place <strong>of</strong> oral intraepi<strong>the</strong>lialneoplasia (OIN). Oral Oncol 38:125–130201. Kui LL, Xiu HZ, Ning LY (2003) Condyloma acuminatum<strong>and</strong> human papilloma virus infection in <strong>the</strong> oral mucosa <strong>of</strong>children. Pediatr Dent 25:149–153202. Lam KY, Law S, Luk JM, Wong J (2001) Oesophageal basaloidsquamous cell carcinoma: a unique clinicopathological entitywith telomerase activity as a prognostic indicator. J Pathol195:435–442203. Lambert PR, Ward PH, Berci G (1980) Pseudosarcoma <strong>of</strong><strong>the</strong> larynx: a comprehensive analysis. Arch Otolaryngol106:700–708204. Lee JJ, Hong WK, Hittelman WN, Mao L, Lotan R, Shin DM,Benner SE, Xu XC, Lee JS, Papadimitrakopolou VM, GeyerC, Perez C, Martin JW, El-Naggar AK, Lippman SM (2000)Predicting cancer development in oral leukoplakia: ten years<strong>of</strong> translational research. Clin Cancer Res 6:1702–1710205. Leifer C, Miller AS, Putong PB, Min BH (1974) Spindle cellcarcinoma <strong>of</strong> oral mucosa: a light <strong>and</strong> electron microscopicstudy <strong>of</strong> apparent sarcomatous metastasis to cervical lymphnodes. Cancer 34:597–605206. Lele SM, Pou AM, Ventura K, Gatalica Z, Payne D (2002)Molecular events in <strong>the</strong> progression <strong>of</strong> recurrent respiratorypapillomatosis to carcinoma. Arch Pathol Lab Med 126:1184–1188207. León X, Quer M, Diez S, Orús C, López-Pousa A, Burgués J(1998) Second neoplasm in patients with head <strong>and</strong> neck cancer.<strong>Head</strong> <strong>Neck</strong> 21:204–210208. León X, Quer M, Orús C, del Prado Venegas M, López M(2000) Distant metastases in head <strong>and</strong> neck cancer patientswho achieved loco-regional control. <strong>Head</strong> <strong>Neck</strong> 22:680–688209. Leong AS, Brown JH (1984) Malignant transformation in athymic cyst. Am J Surg Pathol 8:471–475210. Leventon GS, Evans HL (1981) Sarcomatoid squamous cellcarcinoma <strong>of</strong> <strong>the</strong> mucous membranes <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck: aclinicopathologic study <strong>of</strong> 20 cases. Cancer 48:994–1003211. Lever WF (1947) Adenoacanthoma <strong>of</strong> sweat gl<strong>and</strong>s: carcinoma<strong>of</strong> sweat gl<strong>and</strong>s with gl<strong>and</strong>ular <strong>and</strong> epidermal elements.Report <strong>of</strong> 4 cases. Arch Dermatol Syphilol 56:157–171212. Levi JE, Delcelo R, Alberti VN, Torloni H, Villa LL (1989)Human papillomavirus DNA in respiratory papillomatosisdetected by in situ hybridization <strong>and</strong> <strong>the</strong> polymerase chainreaction. Am J Pathol 135:1179–1184213. Lewis JE, Olsen KD, Sebo TJ (1997) Spindle cell carcinoma<strong>of</strong> <strong>the</strong> larynx: review <strong>of</strong> 26 cases including DNA content <strong>and</strong>immunohistochemistry. Hum Pathol 28:664–673213a. Lewis Jr JS, Ritter JH, El-M<strong>of</strong>ty S (2005) Alternative epi<strong>the</strong>lialmarkers in sarcomatoid carcinomas <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck,lung <strong>and</strong> bladder – p63, MOC-31, <strong>and</strong> TT-F1. Modern <strong>Pathology</strong>18:1471–1481214. Li W, Thompson CH, O’Brien CJ, McNeil EB, Scolyer RA,Cossart YE, Veness MJ, Walker DM, Morgan GJ, Rose BR(2003) Human papillomavirus positivity predicts favourableoutcome for squamous carcinoma <strong>of</strong> <strong>the</strong> tonsil. Int J Cancer106:553–558215. Li WW, Pettit TH, Zakka KA (1980) Intraocular invasion bypapillary squamous cell carcinoma <strong>of</strong> <strong>the</strong> conjunctiva. Am JOphthalmol 90:697–701216. Lindeberg H, Elbrond O (1989) Laryngeal papillomas: clinicalaspects in a series <strong>of</strong> 231 patients. Clin Otolaryngol14:333–342217. Lindeberg H, Elbrond O (1990) Laryngeal papillomas: <strong>the</strong>epidemiology in a Danish subpopulation 1965–1984. ClinOtolaryngol 15:125–131218. Lindeberg H, Elbrond O (1991) Malignant tumours in patientswith a history <strong>of</strong> multiple laryngeal papillomas: <strong>the</strong>significance <strong>of</strong> irradiation. Clin Otolaryngol 16:149–151219. Lindeberg H, Krogdahl A (1997) Laryngeal dysplasia <strong>and</strong> <strong>the</strong>human papillomavirus. Clin Otolaryngol 22:382–386220. Liotta LA (1992) Cancer cell invasion <strong>and</strong> metastasis. Sci Am266:54–59; 62–63221. Liotta LA, Rao CN, Barsky SH (1983) Tumor invasion <strong>and</strong><strong>the</strong> extracellular matrix. Lab Invest 49:636–649222. Luna MA, El Naggar A, Parichatikanond P, Weber SR, BatsakisJG (1990) Basaloid squamous carcinoma <strong>of</strong> <strong>the</strong> upperaerodigestive tract. Clinicopathologic <strong>and</strong> DNA flow cytometricanalysis. Cancer 66:537–542223. Luna MA, Pineda-Daboin K (2001) Upper aerodigestivetract. In: Henson <strong>and</strong> Albores-Savedra’s <strong>Pathology</strong> <strong>of</strong> incipientneoplasia, 3rd edn. Oxford, New York, pp 57–85224. Lundgren JAV, van Nostr<strong>and</strong> AWP, Harwood AR, Cullen RJ,Bryce DP (1986) Verrucous carcinoma (Ackermans’s tumor)<strong>of</strong> <strong>the</strong> larynx: diagnostic <strong>and</strong> <strong>the</strong>rapeutic considerations.<strong>Head</strong> <strong>Neck</strong> Surg 9:19–26225. Luzar B, Poljak M, Marin IJ, Eberlinc A, Klopčič U, Gale N(2004) Human telomerase catalytic subunit gene re-expressionis an early event in oral carcinogenesis. Histopathology45:13–19226. Luzar B, Poljak M, Marin IJ, Gale N (2003) Telomerase reactivationis an early event in laryngeal carcinogenesis. ModPathol 16:841–848227. MacMillan C, Kapadia SB, Finkelstein SD, Nalesnik MA,Barnes L (1996) Lymphoepi<strong>the</strong>lial carcinoma <strong>of</strong> <strong>the</strong> larynx<strong>and</strong> hypopharynx: study <strong>of</strong> eight cases with relationship toEpstein-Barr virus <strong>and</strong> p53 gene alterations, <strong>and</strong> review <strong>of</strong><strong>the</strong> literature. Hum Pathol 27:1172–1179228. Maier H, Tisch M (1997) Epidemiology <strong>of</strong> laryngeal cancer:results <strong>of</strong> <strong>the</strong> Heidelberg case-control study. Acta OtolaryngolSuppl 527:160–164229. Mall<strong>of</strong>ré C, Cardesa A, Campo E, Condom E, Palacin A, Garin-ChesaP, Traserra J (1993) Expression <strong>of</strong> cytokeratins insquamous cell carcinomas <strong>of</strong> <strong>the</strong> larynx: immunohistochemicalanalysis <strong>and</strong> correlation with prognostic factors. PatholRes Pract 189:275–282230. Mamelle G, Pampurik J, Luboinski B, Lancar R, Lusinchi A,Bosq J (1994) Lymph node prognostic factors in head <strong>and</strong>neck squamous cell carcinomas. Am J Surg 168:494–498231. Mao L (1997) Leukoplakia: molecular underst<strong>and</strong>ing <strong>of</strong> premalignantlesions <strong>and</strong> implications for clinical management.Mol Med Today 3:442–448232. Martin MR, Kahn LB (1977) So-called pseudosarcoma <strong>of</strong><strong>the</strong> esophagus: nodal metastases <strong>of</strong> <strong>the</strong> spindle cell element.Arch Pathol Lab Med 101:604–609233. Martinez-Gimeno C, Rodriguez EM, Villa CN, Varela CL(1995) Squamous carcinoma <strong>of</strong> <strong>the</strong> oral cavity: a clinicopathologicscoring system for evaluating risk <strong>of</strong> cervical lymphnode metastasis. Laryngoscope 105:107–114234. Martinez-Madrigal E, Baden E, Casiraghi O, Micheau C(1991) Oral <strong>and</strong> pharyngeal adenosquamous carcinoma. Areport <strong>of</strong> four cases with immunohistochemical studies. EurArch Otorhinolaryngol 248:255–258235. Mashberg A, Samit A (1995) Early diagnosis <strong>of</strong> asymptomaticoral <strong>and</strong> oropharyngeal squamous cancers. CA Cancer JClin 45:328–351


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 35236. McCaffrey TV, Witte M, Ferguson MT (1998) Verrucous carcinoma<strong>of</strong> <strong>the</strong> larynx. Ann Otol Rhinol Laryngol 107:391–395237. McDonalds JS, Crissman JD, Gluckman JL (1982) Verrucouscarcinoma <strong>of</strong> <strong>the</strong> oral cavity. <strong>Head</strong> <strong>Neck</strong> Surg 5:22–28238. McKaig RG, Baric RS, Olshan AF (1998) Human papillomavirus<strong>and</strong> head <strong>and</strong> neck cancer. Epidemiology <strong>and</strong> molecularbiology. <strong>Head</strong> <strong>Neck</strong> 20:250–265239. McLaren KM, Burnett RA, Goodl<strong>and</strong> JR, Howatson SR,Lang S, Lee FD, Lessells AM, Ogston S, Robertson AJ, SimpsonJG, Smith GD, Tavadia HB, Walker F; The Scottish <strong>Pathology</strong>Consistency Group (2000) Consistency <strong>of</strong> histopathologicalreporting <strong>of</strong> laryngeal dysplasia. Histopathology37:460–463240. Medina JE, Dichtel W (1984) Verrucous carcinoma <strong>of</strong> <strong>the</strong>head <strong>and</strong> neck. Arch Otolaryngol 110:437–440241. Meijer JW, Ramaekers FC, Manni JJ, Slo<strong>of</strong>f JJ, AldeweireldtJ, Vooys GP (1988) Intermediate filament proteins in spindlecell carcinoma <strong>of</strong> <strong>the</strong> larynx <strong>and</strong> tongue. Acta Otolaryngol106:306–313242. Michaels L, Hellquist HB (2001) Ear, nose <strong>and</strong> throat histopathology,2nd edn. Springer, London, pp 15–51243. Milford CA, O’Flynn PE (1991) Management <strong>of</strong> verrucouscarcinoma <strong>of</strong> <strong>the</strong> larynx. Clin Otolaryngol 16:160–162244. Miller CS, White DK, Royse DD (1993) In situ hybridizationanalysis <strong>of</strong> human papillomavirus in or<strong>of</strong>acial lesions using aconsensus biotinylated probe. Am J Dermatopathol 15:256–259245. Minić AJ, Stajčić Z (1994) Adenosquamous carcinoma <strong>of</strong><strong>the</strong> inferior turbinate: a case report. J Oral Maxill<strong>of</strong>ac Surg52:764–767246. Mohit-Tabatabai MA, Sobel HJ, Rush BF, Mashberg A (1986)Relation <strong>of</strong> thickness <strong>of</strong> floor <strong>of</strong> mouth stage-I <strong>and</strong> stage-IIcancers to regional metastases. Am J Surg 152:351–353247. Moreno-Lopez LA, Esparza-Gomez GC, Gonzalez-NavarroA, Cerero-Lapiedra R, Gonzalez-Hern<strong>and</strong>ez MJ, Dominguez-RojasV (2000) Risk <strong>of</strong> oral cancer associated with tobaccosmoking, alcohol consumption <strong>and</strong> oral hygiene: acase-control study in Madrid, Spain. Oral Oncol 36:170–174248. Morice WG, Ferreiro JA (1998) Distinction <strong>of</strong> basaloid squamouscell carcinoma from adenoid cystic <strong>and</strong> small cell undifferentiatedcarcinoma by immunohistochemistry. HumPathol 29:609–612249. Műller KM, Krohn RB (1980) Smoking habits <strong>and</strong> <strong>the</strong>ir relationshipto precancerous lesions <strong>of</strong> <strong>the</strong> larynx. J Cancer ResClin Oncol 96:211–217250. Muller S, Barnes L (1995) Basaloid squamous cell carcinoma<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck with a spindle cell component. An unusualhistologic variant. Arch Pathol Lab Med 119:181–182251. Munck-Wikl<strong>and</strong> E, Edström S, Jungmark E, KuylenstiernaR, Lindholm J, Auer G (1994) Nuclear DNA content, proliferating-cellnuclear antigen (PCNA) <strong>and</strong> p53 immunostainingin predicting progression <strong>of</strong> laryngeal cancer in situ lesions.Int J Cancer 56:95–99252. Muscat JE, Wynder EL (1992) Tobacco, alcohol, asbestos,<strong>and</strong> occupational risk factors for laryngeal cancer. Cancer69:2244–2251253. Myers EN, Alvi N (1996) Management <strong>of</strong> <strong>the</strong> carcinoma <strong>of</strong><strong>the</strong> supraglottic larynx: evolution, current concepts, <strong>and</strong> futuretrends. Laryngoscope 106:559–567254. Myssiorek D, Vambutas A, Abramson AL (1994) Carcinomain situ <strong>of</strong> <strong>the</strong> glottic larynx. Laryngoscope 104:463–467255. Nadal A, Campo E, Pinto J, Mall<strong>of</strong>re C, Palacin A, Arias C,Traserra J, Cardesa A (1995) p53 expression in normal, dysplastic,<strong>and</strong> neoplastic laryngeal epi<strong>the</strong>lium. Absence <strong>of</strong> acorrelation with prognostic factors. J Pathol 175:181–188256. Nadal A, Cardesa A (2003) Molecular biology <strong>of</strong> laryngealsquamous cell carcinoma. Virchows Arch 442:1–7257. Nadal A, Jares P, Cazorla M, Fernández PL, Sanjuan X,Hernández L, Pinyol M, Aldea M, Mall<strong>of</strong>ré C, Muntané J,Traserra J, Campo E, Cardesa A (1997) p21WAF1/Cip1 expressionis associated with cell differentiation but not withp53 mutations in squamous cell carcinomas <strong>of</strong> <strong>the</strong> larynx. JPathol 183:156–163258. Napier SS, Gormley JS, Newl<strong>and</strong>s C, Ramsay-Baggs P (1995)Adenosquamous carcinoma. A rare neoplasm with an aggressivecourse. Oral Surg Oral Med Oral Pathol Oral RadiolEndod 79:607–611259. Nappi O, Pettinato G, Wick MR (1989) Adenoid (acantholytic)squamous cell carcinoma <strong>of</strong> <strong>the</strong> skin. J Cutan Pathol16:114–121260. Nappi O, Wick MR, Pettinato G, Ghiselli RW, Swanson PE(1992) Pseudovascular adenoid squamous cell carcinoma <strong>of</strong><strong>the</strong> skin. A neoplasm that may be mistaken for angiosarcoma.Am J Surg Pathol 16:429–438261. Nathan CAO, S<strong>and</strong>ers K, Abreo FW, Nassar R, Glass J (2000)Correlation <strong>of</strong> p53 <strong>and</strong> <strong>the</strong> proto-oncogene eIF4E in larynxcancers: prognostic implications. Cancer Res 60:3599–3604262. Naunheim KS, Taylor JR, Skosey C, H<strong>of</strong>fman PC, FergusonMK, Golomb HM, Little AG (1987) Adenosquamous lungcarcinoma, clinical characteristics, treatment <strong>and</strong> prognosis.Ann Thorac Surg 44:462–466263. Neville BW, Day TA (2002) Oral cancer <strong>and</strong> precancerous lesions.CA Cancer J Clin 52:195–215264. Newman J, Antonakopoulos GN, Darnton J, Mat<strong>the</strong>ws HR(1992) The ultrastructure <strong>of</strong> oesophageal carcinoma: multidirectionaldifferentiation. A transmission microscope study<strong>of</strong> 43 cases. J Pathol 167:192–198265. Niedobitek G, Hansmann ML, Herbst H, Young LS, DienemannD, Hartmann CA et al. (1991) Epstein-Barr-virus <strong>and</strong>carcinomas – undifferentiated carcinomas but not squamous-cellcarcinomas <strong>of</strong> <strong>the</strong> nasopharynx are regularly associatedwith <strong>the</strong> virus. J Pathol 165:17–24266. Nishijima W, Takooda S, Tokita N, Takayama S, Sakura M(1993) Analyses <strong>of</strong> distant metastases in squamous cell carcinoma<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck <strong>and</strong> lesions above <strong>the</strong> clavicles atautopsy. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 119:65–68267. Nunez DA, Astley SM, Lewis FA, Wells M (1994) Humanpapilloma viruses: a study <strong>of</strong> <strong>the</strong>ir prevalence in <strong>the</strong> normallarynx. J Laryngol Otol 108:319–320268. Oijen MCGT van, Leppers vd Straat FGJ, Tilanus MGJ, SlootwegPJ (2000) The origin <strong>of</strong> multiple squamous cell carcinomasin <strong>the</strong> aerodigestive tract. Cancer 88:884–893269. Oijen MG van, Rijksen G, ten Broek FW, Slootweg PJ (1998)Increased expression <strong>of</strong> epidermal growth factor receptors innormal epi<strong>the</strong>lium adjacent to head <strong>and</strong> neck carcinomas independent<strong>of</strong> tobacco <strong>and</strong> alcohol abuse. Oral Dis 4:4–8270. Oijen MGCT van, Slootweg PJ (2000) Gain-<strong>of</strong>-function mutationsin <strong>the</strong> tumor suppressor gene p53. Clin Cancer Res6:2138–2145271. Oijen MGCT van, Slootweg PJ (2000) Review. Oral field cancerization.Carcinogen-induced independent events or micrometastaticdeposits? Cancer Epidemiol Biomarkers Prev9:249–256272. Oijen MGCT van, Tilanus MG, Medema RH, Slootweg PJ(1998) Expression <strong>of</strong> p21 (Waf1/Cip1) in head <strong>and</strong> neck cancerin relation to proliferation, differentiation, p53 status. JOral Pathol Med 27:367–375273. Onishi A, Nakashiro K, Mihara M, Sumida T, Kawamata H,Shintani S, Aida T, Tachikawa T, Kamakawa H (2004) Basic<strong>and</strong> clinical studies on quantitative analysis <strong>of</strong> lymph nodemicrometastasis in oral cancer. Oncol Rep 11:33–39274. Orvidas LJ, Kerry DK, Lewis JE, Suman VJ (1998) Verrucouscarcinoma <strong>of</strong> <strong>the</strong> larynx. <strong>Head</strong> <strong>Neck</strong> 20:197–203275. O’Sullivan B, Warde P, Keane T, Irish J, Cummings B, PayneD (1995) Outcome following radio<strong>the</strong>rapy in verrucous carcinoma<strong>of</strong> <strong>the</strong> larynx. Int J Radiat Oncol Biol Phys 32:611–617276. Parnes SM (1990) Asbestos <strong>and</strong> cancer <strong>of</strong> <strong>the</strong> larynx: is <strong>the</strong>rea relationship? Laryngoscope 100:254–261277. Paulino AFG, Singh B, Shah JP, Huvos AG (2000) Basaloidsquamous cell carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Laryngoscope110:1479–1482278. Perez CA, Kraus FT, Evans JC, Powers WE (1966) Anaplastictransformation in verrucous carcinoma <strong>of</strong> <strong>the</strong> oral cavity afterradiation <strong>the</strong>rapy. Radiology 86:108–115279. Piattelli A, Rubini C, Fioroni M, Iezzi G, Santinelli A (2002)Prevalence <strong>of</strong> p53, bcl-2, <strong>and</strong> Ki-67 immunoreactivity <strong>and</strong>


36 N. Gale · N. Zidar1<strong>of</strong> apoptosis in normal oral epi<strong>the</strong>lium <strong>and</strong> in premalignant<strong>and</strong> malignant lesions <strong>of</strong> <strong>the</strong> oral cavity. J Oral Maxill<strong>of</strong>acSurg 60:532–540280. Pinsolle J, Pinsolle V, Majoufre C, Duroux S, Demeaux H, SiberchicotF (1997) Prognostic value <strong>of</strong> histologic findings inneck dissections for squamous cell carcinoma. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 123:145–148281. Poljak M, Gale N, Kambič V (1997) Human papilloma viruses:a study <strong>of</strong> <strong>the</strong>ir prevalence in <strong>the</strong> epi<strong>the</strong>lial hyperplasticlesions <strong>of</strong> <strong>the</strong> larynx. Acta Otolaryngol Suppl 527:66–69282. Poljak M, Gale N, Kambič V, Ferluga D, Fischinger J (1996)Overexpression <strong>of</strong> p53 protein in benign <strong>and</strong> malignant laryngealepi<strong>the</strong>lial lesions. Anticancer Res 16:1947–1951283. Poljak M, Seme K, Gale N (1998) Detection <strong>of</strong> human papillomavirusesin tissue specimens. Adv Anat Pathol 5:216–234284. Pou AM, Rimell FL, Jordan JA, Shoemaker DL, Johnson JT,Barua P, Post JC, Ehrlich GD (1995) Adult respiratory papillomatosis:human papillomavirus type <strong>and</strong> viral coinfectionsas predictors <strong>of</strong> prognosis. Ann Otol Rhinol Laryngol104:758–762285. Praetorius F (1997) HPV-associated diseases <strong>of</strong> oral mucosa.Clin Dermatol 15:399–413286. Premoli-De-Percoco G, Cisternas JP, Ramirez JL, Galindo I(1993) Focal epi<strong>the</strong>lial hyperplasia: human-papillomavirusinduceddisease with a genetic predisposition in a Venezuelanfamily. Hum Genet 91:386–388287. Pr<strong>of</strong>fitt SD, Spooner TR, Kosek JC (1970) Origin <strong>of</strong> undifferentiatedneoplasm from verrucous epidermal carcinoma <strong>of</strong>oral cavity following irradiation. Cancer 26:389–393288. Quick CA, Foucar E, Dehner LP (1979) Frequency <strong>and</strong> significance<strong>of</strong> epi<strong>the</strong>lial atypia in laryngeal papillomatosis. Laryngoscope89:550–560289. Quiney RE, Wells M, Lewis FA, Terry RM, Michaels L, Cr<strong>of</strong>tCB (1989) Laryngeal papillomatosis: correlation between severity<strong>of</strong> disease <strong>and</strong> presence <strong>of</strong> HPV 6 <strong>and</strong> 11 detected by insitu DNA hybridisation. J Clin Pathol 42:694–698290. Rady PL, Schnadig VJ, Weiss RL, Hughes TK, Tyring SK(1998) Malignant transformation <strong>of</strong> recurrent respiratorypapillomatosis associated with integrated human papillomavirustype 11 DNA <strong>and</strong> mutation <strong>of</strong> p53. Laryngoscope108:735–740291. Rafferty MA, O’Dwyer TP (2001) Secondary primary malignanciesin head <strong>and</strong> neck squamous cell carcinoma. J LaryngolOtol 115:988–991292. R<strong>and</strong>all ME, Andersen WA, Mills SE, Kim JA (1986) Papillarysquamous cell carcinoma <strong>of</strong> uterine cervix: a clinicopathologicstudy <strong>of</strong> nine cases. Int J Gynecol Pathol 5:1–10293. Raslan WF, Barnes L, Krause JR, Contis L, Killeen R, KapadiaSB (1994) Basaloid squamous cell carcinoma <strong>of</strong> <strong>the</strong> head<strong>and</strong> neck: a clinicopathologic <strong>and</strong> flow cytometric study <strong>of</strong> 10new cases with review <strong>of</strong> <strong>the</strong> English literature. Am J Otolaryngol15:204–211294. Regaud C, Reverchon L (1921) Sur un cas d’epi<strong>the</strong>lioma epidermoidedeveloppé dans le massif maxillaire supérieureétendu aux ligaments de la face, aux cavaties buccale, nasaleet orbitaire ainsi que aux ganglions du cou guéri par la radiothérapie.Rev Laryngol Otol Rhinol (Bord) 42:369–378295. Regezi JA, Sciubba JJ, Jordan RCK (2003) Oral pathology.Clinical pathologic correlations, 4th edn. Saunders, St. Louis,pp 143–156296. Rehberg E, Kleinsasser O (1999) Malignant transformationin non-irradiated juvenile laryngeal papillomatosis. EurArch Otorhinolaryngol 256:450–454297. Reibel J (2003) Prognosis <strong>of</strong> oral pre-malignant lesions: significance<strong>of</strong> clinical, histopathological, <strong>and</strong> molecular biologicalcharacteristics. Crit Rev Oral Biol Med 14:47–62298. Reichart PA (2001) Identification <strong>of</strong> risk groups for oral precancer<strong>and</strong> cancer <strong>and</strong> preventive measures. Clin Oral Investig5:207–213299. Reid BC, Winn DM, Morse DE, Pendrys DG (2000) <strong>Head</strong><strong>and</strong> neck in situ carcinoma: incidence, trends, <strong>and</strong> survival.Oral Oncol 36:414–420300. Renan MJ (1993) How many mutations are required for tumorigenesis?Implications from human cancer data. MolCarcinog 7:139–146301. Rihkanen H, Aaltonen LM, Syrjanen SM (1993) Human papillomavirusin laryngeal papillomas <strong>and</strong> in adjacent normalepi<strong>the</strong>lium. Clin Otolaryngol 18:470–474302. Rihkanen H, Peltomaa J, Syrjänen S (1994) Prevalence <strong>of</strong> humanpapillomavirus DNA in vocal cords without laryngealpapillomas. Acta Otolaryngol 114:348–351303. Rindum JL, Stenderup A, Holmstrup P (1994) Identification<strong>of</strong> C<strong>and</strong>ida albicans types related to healthy <strong>and</strong> pathologicaloral mucosa. J Oral Pathol Med 23:406–411304. Rizzardi C, Frezzini C, Maglione M, Tirelli G, Melato M(2003) A look at <strong>the</strong> biology <strong>of</strong> spindle cell squamous carcinoma<strong>of</strong> <strong>the</strong> oral cavity: report <strong>of</strong> a case. J Oral Maxill<strong>of</strong>acSurg 61:264–268305. Rosen PP, Ernsberger D (1987) Low-grade adenosquamouscarcinoma. A variant <strong>of</strong> metaplastic mammary carcinoma.Am J Surg Pathol 11:351–358306. Rosin MP, Cheng X, Poh C, Lam WL, Huang Y, Lovas J, BereanK, Epstein JB, Priddy R, Le ND, Zhang L (2000) Use <strong>of</strong> allelicloss to predict malignant risk for low-grade oral epi<strong>the</strong>lialdysplasia. Clin Cancer Res 6:357–362307. Ross GL, Soutar DS, MacDonald DG, Shoaib T, Camilleri IG,Robertson AG (2004) Improved staging <strong>of</strong> cervical metastasesin clinically node-negative patients with head <strong>and</strong> necksquamous cell carcinoma. Ann Surg Oncol 11:213–218308. Rotfield RE, Myers EN, Johnson JT (1991) Carcinoma in situ<strong>and</strong> microinvasive squamous cell carcinoma <strong>of</strong> <strong>the</strong> vocalcord. Ann Otol Rhinol Laryngol 100:793–796309. Ryan RE, de Santo LW, Devine KD, Weil<strong>and</strong> LH (1977) Verrucouscarcinoma <strong>of</strong> <strong>the</strong> larynx. Laryngoscope 87:1989–1994310. Saito R, Davis BK, Ollapathy EP (1984) Adenosquamous carcinoma<strong>of</strong> <strong>the</strong> prostate. Hum Pathol 15:87–89311. Sakakura A, Yamamoto Y, Takasaki T, Makimoto K, NakamuraM, Takahashi H (1996) Recurrent laryngeal papillomatosisdeveloping into laryngeal carcinoma with human papillomavirus (HPV) type 18: a case report. J Laryngol Otol110:75–77312. Saltarelli MG, Fleming MV, Wenig BM, Gal AA, MansourKA, Travis WD (1995) Primary basaloid squamous cell carcinoma<strong>of</strong> <strong>the</strong> trachea. Am J Clin Pathol 104:594–598313. S<strong>and</strong>erson RJ, Rivron PR, Wallace WA (1991) Adenosquamouscarcinoma <strong>of</strong> <strong>the</strong> hypopharynx. J Laryngol Otol105:678–680314. Scheifele C, Reichart PA, Dietrich T (2003) Low prevalence <strong>of</strong>oral leukoplakia in a representative sample <strong>of</strong> <strong>the</strong> US population.Oral Oncol 39:619–625315. Schmincke A (1921) Über lympho-epi<strong>the</strong>liale Geschwülste.Beitr Pathol Anat 68:161–170316. Sciubba JJ (1995) Oral leukoplakia. Crit Rev Oral Biol Med6:147–160317. Scully C, Porter SR, Speight PM, Eveson JW, Gale D (1999)Adenosquamous carcinoma <strong>of</strong> <strong>the</strong> mouth: a rare variant <strong>of</strong>squamous cell carcinoma. Int J Oral Maxill<strong>of</strong>ac Surg 28:125–128318. Seidman JD, Berman JJ, Yost BA, Iseri OA (1991) Basaloidsquamous carcinoma <strong>of</strong> <strong>the</strong> hypopharynx <strong>and</strong> larynx associatedwith second primary tumors. Cancer 68:1545–1549319. Shafer WG, Waldron CA (1975) Erythroplakia <strong>of</strong> <strong>the</strong> oralcavity. Cancer 36:1021–1028320. Shah JP (1990) Patterns <strong>of</strong> cervical lymph node metastasesfrom squamous carcinomas <strong>of</strong> <strong>the</strong> upper aerodigestive tract.Am J Surg 160:405–409321. Shah KV, Stern WF, Shah FK, Bishai D, Kashima HK (1998)Risk factors for juvenile onset recurrent respiratory papillomatosis.Pediatr Infect Dis J 17:372–376322. Silverman S Jr, Gorsky M (1997) Proliferative verrucous leukoplakia:a follow-up study <strong>of</strong> 54 cases. Oral Surg Oral MedOral Pathol Oral Radiol Endod 84:154–157323. Silverman S Jr, Gorsky M, Lozada F (1984) Oral leukoplakia<strong>and</strong> malignant transformation. A follow-up study <strong>of</strong> 257 patients.Cancer 53:563–568


Lesions <strong>of</strong> Squamous Epi<strong>the</strong>lium Chapter 1 37324. Slaughter DP, Southwick HW, Smejkal W (1953) “Field cancerization”in oral stratified squamous epi<strong>the</strong>lium: clinicalimplication <strong>of</strong> multicentric origin. Cancer 6:963–968325. Slootweg PJ, Hordijk GJ, Koole R (1996) Autopsy findings inpatients with head <strong>and</strong> neck squamous cell cancer <strong>and</strong> <strong>the</strong>ir<strong>the</strong>rapeutic relevance. Oral Oncol Eur J Cancer 32B:413–415326. Slootweg PJ, Hordijk GJ, Schade Y, Es RJJ van, Koole R (2002)Treatment failure <strong>and</strong> margin status in head <strong>and</strong> neck cancer.A critical view on <strong>the</strong> potential value <strong>of</strong> molecular pathology.Oral Oncol 38:500–503327. Slootweg PJ, Muller H (1983) Verrucous hyperplasia or verrucouscarcinoma. An analysis <strong>of</strong> 27 patients. J Maxill<strong>of</strong>acSurg 11:13–19328. Slootweg PJ, Richardson M (2001) Squamous cell carcinoma<strong>of</strong> <strong>the</strong> upper aerodigestive system. In: Gnepp DR (ed) Diagnosticsurgical pathology <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Saunders,Philadelphia, pp 19–78329. Slootweg PJ, Roholl PJM, Müller H et al. (1987) Spindle cellcarcinoma <strong>of</strong> <strong>the</strong> oral cavity <strong>and</strong> larynx. Immunohistochemicalaspects. J Craniomaxfac Surg 17:234–236330. Smith EM, Pignatari SS, Gray SD, Haugen TH, Turek LP(1993) Human papillomavirus infection in papillomas <strong>and</strong>nondiseased respiratory sites <strong>of</strong> patients with recurrent respiratorypapillomatosis using <strong>the</strong> polymerase chain reaction.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 119:554–557331. Smith EM, Summersgill KF, Allen J, H<strong>of</strong>fman HT, Mc-Culloch T, Turek LP, Haugen TH (2000) Human papillomavirus<strong>and</strong> risk <strong>of</strong> laryngeal cancer. Ann Otol Rhinol Laryngol109:1069–1076332. Sobin LH, Wittekind C (2002) TNM: classification <strong>of</strong> malignanttumours, 6th edn. Wiley, New York333. Soo KC, Carter RL, O‘Brien CJ, Barr L, Bliss JM, Shaw HJ(1986) Prognostic implication <strong>of</strong> perineural spread in squamouscarcinomas <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Laryngoscope96:1145–1148334. Sood S, O’Hara J, Quraishi HS (2002) The significance <strong>of</strong> oralleukoplakia. Curr Opin Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 10:80–84335. Souglu Y, Erdamar B, Katircioglu OS, Karatay MC, Sunay T(2002) Extracapsular spread in ipsilateral neck <strong>and</strong> contralateralneck metastases in laryngeal cancer. Ann Otol RhinolLaryngol 111:447–454336. Spayne JA,Warde P, O‘Sullivan B, Payne D, Liu FF, WaldronJ, Gullane PJ, Cummings BJ (2001) Carcinoma in situ <strong>of</strong> <strong>the</strong>glottic larynx: results <strong>of</strong> <strong>the</strong> treatment with radiation <strong>the</strong>rapy.Int J Radiat Oncol Biol Phys 49:1235–1238337. Spector JG, Sessions DG, Haughey BH, Chao KSC, SimpsonJ, Al M<strong>of</strong>ty S, Perez CA (2001) Delayed regional metastases,distant metastases, <strong>and</strong> second primary malignancies insquamous cell carcinomas <strong>of</strong> <strong>the</strong> larynx <strong>and</strong> hypopharynx.Laryngoscope 111:1079–1087338. Sperry K (1994) Lethal asphyxiating juvenile laryngeal papillomatosis.Am J Forensic Med Pathol 15:146–150339. Spiro RH (1998) Verrucous carcinoma, <strong>the</strong>n <strong>and</strong> now. Am JSurg 176:393–397340. Staley CJ, Ujiki GT, Yokoo H (1977) “Pseudocarcinoma” <strong>of</strong><strong>the</strong> larynx: independent metastasis <strong>of</strong> carcinomatous <strong>and</strong>sarcomatous elements. Arch Otolaryngol 94:458–465341. Stutsman AC, McGavran MH (1971) Ultraconservative management<strong>of</strong> superficially invasive epidermoid carcinoma <strong>of</strong><strong>the</strong> true vocal cord. Ann Otol Rhinol Laryngol 80:506–512342. Suarez P, Batsakis JG, el-Naggar AK (1998) Leukoplakia: stilla gallimaufry or is progress being made? A review. Adv AnatPathol 5:137–155343. Suarez PA, Adler-Storthz K, Luna MA, El-Naggar AK, Abdul-KarimFW, Batsakis JG (2000) Papillary squamous cellcarcinoma <strong>of</strong> <strong>the</strong> upper aerodigestive tract: a clinicopathologic<strong>and</strong> molecular study. <strong>Head</strong> <strong>Neck</strong> 22:60–368344. Sudbo J, Kildal W, Risberg B, Koppang HS, Danielsen HE,Reith A (2001) DNA content as a prognostic marker in patientswith oral leukoplakia. N Engl J Med 344:1270–1278345. Sugiyama M, Bhawal UK, Dohmen T, Ono S, Miyauchi M,Ishikawa T (2003) Detection <strong>of</strong> human papillomavirus-16<strong>and</strong> HPV-18 DNA in normal, dysplastic, <strong>and</strong> malignant oralepi<strong>the</strong>lium. Oral Surg Oral Med Oral Pathol Oral Radiol Endod95:594–600346. Syrjänen K, Syrjänen S (2000) Papillomavirus infections inhuman pathology. Wiley, Chichester, pp 1–615347. Tabor MP, Houten VMM van, Kummer JA, Vosjan MJWD,Vlasblom R, Snow GB, Leemans CR, Braakhuis BJM, Brakenh<strong>of</strong>fRH (2002) Discordance <strong>of</strong> genetic alterations betweenprimary head <strong>and</strong> neck tumors <strong>and</strong> corresponding metastasesassociated with mutational status <strong>of</strong> <strong>the</strong> TP53gene. GenesChromosomes Cancer 33:168–177348. Takagi M, Sakota Y, Takayama S, Ishikawa G (1977) Adenoidsquamous cell carcinoma <strong>of</strong> <strong>the</strong> oral mucosa. Report <strong>of</strong> twoautopsy cases. Cancer 40:2250–2255349. Takata T, Ito H, Ogawa I, Miyaguchi M, Iljuhin N, Nikai H(1991) Spindle cell squamous carcinoma <strong>of</strong> <strong>the</strong> oral region.An immunohistochemical <strong>and</strong> ultrastructural study on <strong>the</strong>histogenesis <strong>and</strong> differential diagnosis with a clinicopathologicanalysis <strong>of</strong> six cases. Virchows Arch 419:177–182350. Takes RP, Baatenburg de Jong RJ, van Blommestein R, HermansJ, van Krieken HHJM, Cornelisse CJ (2002) DNA ploidystatus as a prognostic marker <strong>and</strong> predictor <strong>of</strong> lymph nodemetastasis in laryngeal carcinoma. Ann Otol Rhinol Laryngol111:1015–1020351. Tavani A, Negri E, Franceschi S, Barbone F, La Vecchia C(1994) Attributable risk for laryngeal cancer in nor<strong>the</strong>rn Italy.Cancer Epidemiol Biomarkers Prev 3:121–125352. Terry RM, Lewis FA, Robertson S, Bly<strong>the</strong> D, Wells M (1989)Juvenile <strong>and</strong> adult laryngeal papillomata: classification by insituhybridization for human papillomavirus. Clin Otolaryngol14:135–139353. Tharp ME, Shidnia H (1995) Radio<strong>the</strong>rapy in <strong>the</strong> treatment<strong>of</strong> verrucous carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Laryngoscope105:391–396354. Thompson L, Chang B, Barsky SH (1996) Monoclonal origins<strong>of</strong> malignant mixed tumors (carcinosarcomas). Evidence fora divergent histogenesis. Am J Surg Pathol 20:277–285355. Thompson LDR, Wenig BM, Heffner DK, Gnepp DR (1999)Exophytic <strong>and</strong> papillary squamous cell carcinomas <strong>of</strong> <strong>the</strong>larynx: a clinicopathologic series <strong>of</strong> 104 cases. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 120:718–724356. Thompson LDR, Wieneke JA, Miettinen M, Heffner DK(2002) Spindle cell (sarcomatoid) carcinomas <strong>of</strong> <strong>the</strong> larynx:a clinicopathologic study <strong>of</strong> 187 cases. Am J Surg Pathol26:153–170357. Thomson PJ, Soames JV, Booth C, O’Shea JA (2002) Epi<strong>the</strong>lialcell proliferative activity <strong>and</strong> oral cancer progression. CellProlif 1:110–120358. Tjebbes GWA, Kreijveld PA, Tilanus MGJ, Hordijk GJ, SlootwegPJ (2002) P53 tumor suppressor gene mutations in laryngealcancer <strong>and</strong> in recurrent disease following radiation <strong>the</strong>rapy.Oral Oncol 38:296–300359. Tjebbes GWA, Leppers FGJ, Hordijk GJ, Tilanus MGJ, SlootwegPJ (1999) p53 tumor suppressor gene as a clonal markerin head <strong>and</strong> neck squamous cell carcinoma. p53 mutationsin primary tumor <strong>and</strong> matched lymph node metastases. OralOncol 35:384–389360. Toda S, Yonemitsu N, Miyabara S, Sugihara H, Maehara N(1989) Polypoid squamous cell carcinoma <strong>of</strong> <strong>the</strong> larynx. Animmunohistochemical study for ras p21 <strong>and</strong> cytokeratin.Pathol Res Pract 185:860–866361. Toorn PP van der, Veltman JA, Bot FJ, de Jong JM, ManniJJ, Ramaekers FC, Hopman AH (2001) Mapping <strong>of</strong> resectionmargins <strong>of</strong> oral cancer for p53 overexpression <strong>and</strong> chromosomeinstability to detect residual (pre)malignant cells. JPathol 193:66–72362. Traserra J, Comas J, Conde C, Cuchi A, Cardesa A (1990)Metastatic involvement <strong>of</strong> <strong>the</strong> cavernous sinus from primarypharyngolaryngeal tumors. <strong>Head</strong> <strong>Neck</strong> 12:426–429363. Uhlman DL, Adams G, Knapp D, Aeppli MM, Niehans G(1996) Immunohistochemical staining for markers <strong>of</strong> futureneoplastic progression in <strong>the</strong> larynx. Cancer Res 56:2199–2205364. Uzcudun AE, Bravo Fernández P, Sánchez JJ, García Gr<strong>and</strong>eA, Rabanal Retolaza I, González Baron M, Gavilán Bouzas J(2001) Clinical features <strong>of</strong> pharyngeal cancer: a retrospective


38 N. Gale · N. Zidar1study <strong>of</strong> 258 consecutive patients. J Laryngol Otol 115:112–118365. Vaidya MM, Sawant SS, Borges AM, Ogale SB, Bhisey AN(1998) Cytokeratin expression in precancerous lesions <strong>of</strong> <strong>the</strong>human oral cavity. Oral Oncol 34:261–264366. Vedt<strong>of</strong>te P, Holmstrup P, Hjorting-Hansen E, Pindborg JJ(1987) Surgical treatment <strong>of</strong> premalignant lesions <strong>of</strong> <strong>the</strong> oralmucosa. Int J Oral Maxill<strong>of</strong>ac Surg 16:656–664367. Velden LA van der, Schaafsma HE, Manni JJ, Ruiter DJ, RamaekersFCS, Kuijpers W (1997) Cytokeratin expression innormal epi<strong>the</strong>lium <strong>and</strong> squamous cell carcinomas <strong>of</strong> <strong>the</strong> larynx.Eur Arch Otorhinolaryngol 254:376–383368. Violaris NS, O’Neill D, Helliwell TR, Caslin AW, Rol<strong>and</strong> NJ,Jones AS (1994) S<strong>of</strong>t tissue cervical metastases <strong>of</strong> squamouscarcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Clin Otolaryngol 19:394–399369. Volavšek M, Bračko M, Gale N (2003) Distribution <strong>and</strong> prognosticsignificance <strong>of</strong> cell cycle proteins in squamous carcinoma<strong>of</strong> <strong>the</strong> larynx, hypopharynx <strong>and</strong> adjacent epi<strong>the</strong>lial hyperplasticlesions. J Laryngol Otol 117:286–293370. Vural E, Hutcheson J, Korourian S, Kechelava S, Hanna E(2000) Correlation <strong>of</strong> neural cell adhesion molecules withperineural spread <strong>of</strong> squamous cell carcinoma <strong>of</strong> <strong>the</strong> head<strong>and</strong> neck. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 122:717–720371. Waal I van der, Schepman KP, van der Meij EH, Smeele LE(1997) Oral leukoplakia: a clinicopathological review. OralOncol 33:291–301372. Wain SL, Kier R, Volmer RT, Bossen EH (1986) Basaloidsquamouscarcinoma <strong>of</strong> <strong>the</strong> tongue, hypopharynx, <strong>and</strong> larynx.Report <strong>of</strong> 10 cases. Hum Pathol 17:1158–1166373. Waldron CA, Shafer WG. Leukoplakia revisited (1975) Aclinicopathologic study 3256 oral leukoplakias. Cancer36:1386–1392374. Ward KA, Napier SS, Winter PC, Maw RD, Dinsmore WW(1995) Detection <strong>of</strong> human papilloma virus DNA sequencesin oral squamous cell papillomas by <strong>the</strong> polymerase chain reaction.Oral Surg Oral Med Oral Pathol Oral Radiol Endod80:63–66375. Wenig BM (2002) Squamous cell carcinoma <strong>of</strong> <strong>the</strong> upperaerodigestive tract: precursors <strong>and</strong> problematic variants.Mod Pathol 15:229–254376. Wiernik G, Millard PR, Haybittle JL (1991) The predictivevalue <strong>of</strong> histological classification into degrees <strong>of</strong> differentiation<strong>of</strong> squamous cell carcinoma <strong>of</strong> <strong>the</strong> larynx <strong>and</strong> hypopharynxcompared with <strong>the</strong> survival <strong>of</strong> patients. Histopathology19:411–417377. Winzenburg SM, Niehans GA, George E, Daly K, Adams GL(1997) Basaloid squamous carcinoma: a clinical comparison<strong>of</strong> two histologic types with poorly differentiated squamouscell carcinoma. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 119:471–475378. Wolf GT, Fisher SG, Truelson JM, Beals TF (1994) DNA content<strong>and</strong> regional metastases in patients with advanced laryngealsquamous carcinoma. Laryngoscope 104:479–483379. Woolgar JA (1999) Micrometastasis in oral/oropharyngealsquamous cell carcinoma: incidence, histopathological features<strong>and</strong> clinical implications. Br J Oral Maxill<strong>of</strong>ac Surg37:181–186380. Woolgar JA, Rogers SN, Lowe D, Brown JS, Vaughan ED(2003) Cervical lymph node metastasis in oral cancer: <strong>the</strong>importance <strong>of</strong> even microscopic extracapsular spread. OralOncol 39:130–137381. World Health Organization Classification <strong>of</strong> Tumours (2005)<strong>Pathology</strong> <strong>and</strong> genetics <strong>of</strong> tumours <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck.IARC, Lyon382. Yilmaz T, Hoşal AŞ, Gedikoğlu G, Kaya S (1999) Prognosticsignificance <strong>of</strong> histopathological parameters in cancer <strong>of</strong> <strong>the</strong>larynx. Eur Arch Otorhinolaryngol 256:139–144383. Yilmaz T, Hoşal AS, Gedikoĝlu G, Őnerci M, Gürsel B (1998)Prognostic significance <strong>of</strong> vascular <strong>and</strong> perineural invasionin cancer <strong>of</strong> <strong>the</strong> larynx. Am J Otolaryngol 19:83–88384. Yoshihara T, Yamamura Y (1997) An unusual case <strong>of</strong> laryngealcarcinoma metastasizing to <strong>the</strong> small intestine. J LaryngolOtol 111:575–577385. Yoshimura Y, Mishima K, Obara S, Yoshimura H, MaruyamaR (2003) Clinical characteristics <strong>of</strong> oral adenosquamous carcinoma:report <strong>of</strong> a case <strong>and</strong> an analysis <strong>of</strong> <strong>the</strong> reported Japanesecases. Oral Oncol 39:309–315386. Young SK, Min KW (1991) In situ DNA hybridization analysis<strong>of</strong> oral papillomas, leukoplakias, <strong>and</strong> carcinomas forhuman papillomavirus. Oral Surg Oral Med Oral Pathol71:726–729387. Yucel OT, Yilmaz T, Unal OF, Turan E (1999) Distant metastasesin laryngeal squamous cell carcinoma. J Exp Clin CancerRes 18:285–288388. Zaatari GS, Santoianni RA (1986) Adenoid squamous cellcarcinoma <strong>of</strong> <strong>the</strong> nasopharynx <strong>and</strong> neck region. Arch PatholLab Med 110:542–546389. Zain RB (2001) Cultural <strong>and</strong> dietary risk factors <strong>of</strong> oral cancer<strong>and</strong> precancer – a brief overview. Oral Oncol 37:205–210390. Zakrzewska JM, Lopes V, Speight P, Hopper C (1996) Proliferativeverrucous leukoplakia: a report <strong>of</strong> ten cases. OralSurg Oral Med Oral Pathol Oral Radiol Endod 82:396–401391. Zarbo RJ, Crissman JD, Venkat H, Weiss MA (1986) Spindle-cellcarcinoma <strong>of</strong> <strong>the</strong> upper aerodigestive tract mucosa.An immunohistochemical <strong>and</strong> ultrastructural study <strong>of</strong> 18 biphasictumors <strong>and</strong> comparison with seven monophasic spindle-celltumors. Am J Surg Pathol 10:741–753392. Zbären P, Borisch B, Lang H, Greiner R (1997) Undifferentiatedcarcinoma <strong>of</strong> nasopharyngeal type <strong>of</strong> <strong>the</strong> laryngopharyngealregion. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 117:688–693393. Zbären P, Lehmann W (1987) Frequency <strong>and</strong> sites <strong>of</strong> distantmetastases in head <strong>and</strong> neck squamous cell carcinoma. Ananalysis <strong>of</strong> 101 cases at autopsy. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong>Surg 11:762–764394. Zidar N, Gale N, Cör A, Kambič V (1996) Expression <strong>of</strong> Ki-67 antigen <strong>and</strong> proliferative cell nuclear antigen in benign<strong>and</strong> malignant epi<strong>the</strong>lial lesions <strong>of</strong> <strong>the</strong> larynx. J LaryngolOtol 110:440–445395. Zidar N, Gale N, Kambič V, Fischinger J (2001) Expression <strong>of</strong>tenascin <strong>and</strong> fibronectin in benign epi<strong>the</strong>lial hyperplastic lesions<strong>and</strong> squamous carcinoma <strong>of</strong> <strong>the</strong> larynx. Anticancer Res21:451–454396. Zidar N, Gale N, Kambič V, Fischinger J (2002) Proliferation<strong>of</strong> my<strong>of</strong>ibroblasts in <strong>the</strong> stroma <strong>of</strong> epi<strong>the</strong>lial hyperplasticlesions <strong>and</strong> squamous carcinoma <strong>of</strong> <strong>the</strong> larynx. Oncology62:381–385


Chapter 2Nasal Cavity<strong>and</strong> Paranasal SinusesA. Cardesa · L. Alos · A. Franchi2Contents2.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . 402.1.1 Embryology . . . . . . . . . . . . . . . . . . . . . . . 402.1.2 Anatomy . . . . . . . . . . . . . . . . . . . . . . . . . 402.1.3 Histology . . . . . . . . . . . . . . . . . . . . . . . . . 402.2. Acute <strong>and</strong> Chronic Rhinosinusitis . . . . . . . . . . 402.2.1 Viral Infections (Common Cold) . . . . . . . . . . . 402.2.2 Bacterial Infections . . . . . . . . . . . . . . . . . . . 402.2.3 Allergic Rhinitis . . . . . . . . . . . . . . . . . . . . . 402.2.4 Atrophic Rhinitis . . . . . . . . . . . . . . . . . . . . 412.2.5 Hypertrophic Rhinitis . . . . . . . . . . . . . . . . . 412.2.6 Non-Suppurative Chronic Sinusitis . . . . . . . . . . 412.3 Sinonasal Polyps . . . . . . . . . . . . . . . . . . . . . 412.3.1 Allergic Polyposis . . . . . . . . . . . . . . . . . . . . 412.3.2 Polyposis in Mucoviscidosis . . . . . . . . . . . . . . 412.3.3 Polyposis in Immotile Cilia Syndrome<strong>and</strong> in Kartagener’s Syndrome . . . . . . . . . . . . . 412.3.4 Antrochoanal Polyps . . . . . . . . . . . . . . . . . . . 412.4 Sinonasal Hamartomatous<strong>and</strong> Teratoid Lesions . . . . . . . . . . . . . . . . . . 422.4.1 Hamartomas . . . . . . . . . . . . . . . . . . . . . . . 422.4.2 Teratoid Lesions . . . . . . . . . . . . . . . . . . . . . 422.5 Pseudotumours . . . . . . . . . . . . . . . . . . . . . 432.5.1 Mucocele . . . . . . . . . . . . . . . . . . . . . . . . . 432.5.2 Organising Haematoma . . . . . . . . . . . . . . . . 432.5.3 Amyloidosis . . . . . . . . . . . . . . . . . . . . . . . 432.5.4 Myospherulosis . . . . . . . . . . . . . . . . . . . . . 432.5.5 Eosinophilic Angiocentric Fibrosis . . . . . . . . . . 432.5.6 Heterotopic Brain Tissue . . . . . . . . . . . . . . . . 432.6 Fungal Diseases . . . . . . . . . . . . . . . . . . . . . 442.6.1 Aspergillosis . . . . . . . . . . . . . . . . . . . . . . . 442.6.2 Mucormycosis . . . . . . . . . . . . . . . . . . . . . . 442.6.3 Rhinosporidiosis . . . . . . . . . . . . . . . . . . . . 442.7 HIV-Related Infections . . . . . . . . . . . . . . . . . 442.8 Mid-Facial Necrotising Granulomatous Lesions . . 452.8.1 Wegener’s Granulomatosis . . . . . . . . . . . . . . . 452.8.2 Lepromatous Leprosy . . . . . . . . . . . . . . . . . . 452.8.3 Tuberculosis . . . . . . . . . . . . . . . . . . . . . . . 452.8.4 Sarcoidosis . . . . . . . . . . . . . . . . . . . . . . . . 452.8.5 Rhinoscleroma . . . . . . . . . . . . . . . . . . . . . . 452.8.6 Leishmaniasis . . . . . . . . . . . . . . . . . . . . . . 452.8.7 Cocaine Abuse . . . . . . . . . . . . . . . . . . . . . . 462.8.8 Local Steroid Injections . . . . . . . . . . . . . . . . 462.9 Benign Epi<strong>the</strong>lial Neoplasms . . . . . . . . . . . . . 462.9.1 Sinonasal Papillomas . . . . . . . . . . . . . . . . . . 462.9.1.1 Squamous Cell Papilloma . . . . . . . . . . . . . . . 462.9.1.2 Exophytic Papilloma . . . . . . . . . . . . . . . . . . 462.9.1.3 Inverted Papilloma . . . . . . . . . . . . . . . . . . . 462.9.1.4 Oncocytic Papilloma . . . . . . . . . . . . . . . . . . 472.9.2 Salivary-Type Adenomas . . . . . . . . . . . . . . . . 482.9.3 Pituitary Adenomas . . . . . . . . . . . . . . . . . . . 482.10 Benign Sinonasal S<strong>of</strong>t Tissue Neoplasms . . . . . . 482.10.1 Haemangiomas . . . . . . . . . . . . . . . . . . . . . 482.10.2 Haemangiopericytoma . . . . . . . . . . . . . . . . . 482.10.3 Solitary Fibrous Tumour . . . . . . . . . . . . . . . . 482.10.4 Desmoid Fibromatosis . . . . . . . . . . . . . . . . . 492.10.5 Fibrous Histiocytoma . . . . . . . . . . . . . . . . . . 492.10.6 Leiomyoma . . . . . . . . . . . . . . . . . . . . . . . . 492.10.7 Schwannoma <strong>and</strong> Neur<strong>of</strong>ibroma . . . . . . . . . . . 492.10.8 Meningioma . . . . . . . . . . . . . . . . . . . . . . . 502.10.9 Paraganglioma . . . . . . . . . . . . . . . . . . . . . . 502.10.10 Juvenile Angi<strong>of</strong>ibroma . . . . . . . . . . . . . . . . . 502.11 Malignant Sinonasal Tumours . . . . . . . . . . . . 502.11.1 Keratinising Squamous Cell Carcinoma . . . . . . . 512.11.2 Cylindrical Cell Carcinoma . . . . . . . . . . . . . . 522.11.3 Sinonasal Undifferentiated Carcinoma . . . . . . . . 532.11.4 Small Cell (Neuroendocrine) Carcinoma . . . . . . 542.11.5 Primary Sinonasal Nasopharyngeal-TypeUndifferentiated Carcinoma . . . . . . . . . . . . . . 542.11.6 Malignant Melanoma . . . . . . . . . . . . . . . . . . 552.11.7 Olfactory Neuroblastoma . . . . . . . . . . . . . . . 572.11.8 Primitive Neuroectodermal Tumour . . . . . . . . . 582.11.9 High-Grade Sinonasal Adenocarcinomas . . . . . . 582.11.9.1 Intestinal-Type Adenocarcinoma . . . . . . . . . . . 582.11.9.2 Salivary-Type High-Grade Adenocarcinoma . . . . 602.11.10 Low-Grade Sinonasal Adenocarcinomas . . . . . . 602.11.10.1 Non-Salivary-TypeLow-Grade Adenocarcinomas . . . . . . . . . . . . . 602.11.10.2 Salivary-TypeLow-Grade Adenocarcinomas . . . . . . . . . . . . . 612.11.11 Sinonasal Malignant Lymphomas . . . . . . . . . . . 612.11.12 Extramedullary Plasmacytoma . . . . . . . . . . . . 622.11.13 Fibrosarcoma . . . . . . . . . . . . . . . . . . . . . . 622.11.14 Malignant Fibrous Histiocytoma . . . . . . . . . . . 632.11.15 Leiomyosarcoma . . . . . . . . . . . . . . . . . . . . 632.11.16 Rhabdomyosarcoma . . . . . . . . . . . . . . . . . . 632.11.17 Malignant Peripheral Nerve Sheath Tumour . . . . 632.11.18 Teratocarcinosarcoma . . . . . . . . . . . . . . . . . . 63References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64


40 A. Cardesa · L. Alos · A. Franchi22.1 Introduction2.1.1 EmbryologyThe midface, or area between <strong>the</strong> upper lip <strong>and</strong> forehead,develops at between 4 <strong>and</strong> 8 weeks’ gestation [219]. Thefrontal prominence forms during <strong>the</strong> 4th postovulatoryweek <strong>and</strong> gives rise to <strong>the</strong> superior <strong>and</strong> middle portions<strong>of</strong> <strong>the</strong> face. The maxillary <strong>and</strong> nasal swellings form beneath<strong>the</strong> frontal prominence. At <strong>the</strong> end <strong>of</strong> <strong>the</strong> 4th weeksurface thickening <strong>of</strong> <strong>the</strong> nasal swellings forms <strong>the</strong> nasalplacodes, which are <strong>of</strong> ectodermal origin <strong>and</strong> give riseto <strong>the</strong> epi<strong>the</strong>lial lining <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> paranasalsinuses. The placodes invaginate, producing <strong>the</strong> nasalpits that become <strong>the</strong> anterior choanae (nostrils) <strong>and</strong>,less superficially, <strong>the</strong> primitive posterior choanae. Themedial nasal <strong>and</strong> frontal processes give rise to <strong>the</strong> nasalseptum, frontal bones, nasal bones, ethmoid sinus complexes<strong>and</strong> upper incisors. The lateral nasal <strong>and</strong> maxillaryprocesses fuse to form <strong>the</strong> philtrum <strong>and</strong> columella.The cartilaginous nasal capsule forms deep to <strong>the</strong> nasal<strong>and</strong> frontal bones from <strong>the</strong> chondrocranium (skull base)during <strong>the</strong> 7th <strong>and</strong> 8th postovulatory weeks. The paranasalsinuses develop from <strong>the</strong> lateral nasal walls at <strong>the</strong>6th foetal week, <strong>and</strong> <strong>the</strong>ir growth continues after birth,throughout childhood <strong>and</strong> adolescence.2.1.2 AnatomyThe nasal cavities are separated by <strong>the</strong> nasal septum,<strong>and</strong> limited by a ro<strong>of</strong>, which is formed by <strong>the</strong> cribriformplate <strong>of</strong> <strong>the</strong> ethmoid, <strong>and</strong> a floor, which is formed by <strong>the</strong>hard palate [261]. The lateral walls have three turbinatesor conchae, <strong>and</strong> three horizontal spaces, or meatus, oneach side. The nasolacrimal duct opens in <strong>the</strong> inferiormeatus, whereas <strong>the</strong> middle meatus receives drainagefrom <strong>the</strong> frontal, anterior ethmoid <strong>and</strong> maxillary sinuses.Below <strong>the</strong> superior turbinate is <strong>the</strong> sphenoethmoidrecess, with <strong>the</strong> openings <strong>of</strong> <strong>the</strong> sphenoid <strong>and</strong> posteriorethmoid sinuses. Each nasal cavity communicates posteriorlywith <strong>the</strong> nasopharynx through <strong>the</strong> choanae.The paranasal sinuses are a group <strong>of</strong> cavities within <strong>the</strong>corresponding crani<strong>of</strong>acial bones (maxilla, sphenoid,ethmoid <strong>and</strong> frontal) that communicate with <strong>the</strong> nasalcavities through an ostium.2.1.3 HistologyThe nasal vestibule <strong>and</strong> skin share a similar histology.At <strong>the</strong> level <strong>of</strong> <strong>the</strong> limen nasi, <strong>the</strong> keratinising squamousepi<strong>the</strong>lium gradually changes first to cuboidal or columnarepi<strong>the</strong>lium, <strong>and</strong> <strong>the</strong>n to ciliated respiratory-type epi<strong>the</strong>lium,which lines most <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> all <strong>the</strong>paranasal sinuses, with <strong>the</strong> exception <strong>of</strong> <strong>the</strong> ro<strong>of</strong> [261].Numerous goblet cells are interspersed in <strong>the</strong> respiratory-typeepi<strong>the</strong>lium. The lamina propria contains severalseromucous gl<strong>and</strong>s, lymphocytes, monocytes, <strong>and</strong>a well-developed vascular network, particularly evidentin <strong>the</strong> inferior <strong>and</strong> middle turbinate. The olfactory epi<strong>the</strong>liumis predominantly made <strong>of</strong> columnar non-ciliatedsustentacular cells, with scattered bipolar sensoryneurons <strong>and</strong> basal cells.2.2 Acute <strong>and</strong> Chronic Rhinosinusitis2.2.1 Viral Infections (Common Cold)Infectious rhinitis is typically viral <strong>and</strong> is <strong>of</strong>ten referredto as <strong>the</strong> “common cold”. It is more common in childrenthan in adults, <strong>and</strong> <strong>the</strong> most frequently identified agentsare rhinovirus, myxovirus, coronavirus <strong>and</strong> adenovirus[67, 271]. Swelling <strong>of</strong> <strong>the</strong> mucosa may cause obstruction<strong>of</strong> a sinus ostium, with subsequent secondary bacterialinfection (acute bacterial sinusitis). The histologic findingsinclude marked oedema <strong>and</strong> a non-specific mixedinflammatory infiltrate <strong>of</strong> <strong>the</strong> lamina propria.2.2.2 Bacterial InfectionsBacterial rhinosinusitis usually follows a viral infectionor allergic rhinitis, <strong>and</strong> <strong>the</strong> most commonly involvedagents are Streptococcus pneumoniae, Haemophilusinfluenzae <strong>and</strong> Moraxella catarrhalis [11, 34]. A denseinflammatory infiltrate mainly made <strong>of</strong> neutrophils occupies<strong>the</strong> lamina propria. Acute bacterial rhinosinusitisusually resolves with antibiotic <strong>the</strong>rapy. Complicationsare rare <strong>and</strong> include contiguous infectious involvement<strong>of</strong> <strong>the</strong> orbit or central nervous system.2.2.3 Allergic RhinitisAllergic rhinitis (hay fever) is part <strong>of</strong> an inherited syndrome,which may also manifest as atopic eczema <strong>and</strong>asthma. In allergic rhinitis, airborne particles, such asgrass pollens, moulds <strong>and</strong> animal allergens, are depositedon <strong>the</strong> nasal mucosa giving rise to acute <strong>and</strong> chronicreactions. Allergens combine with <strong>the</strong> IgE antibodiesproduced by <strong>the</strong> plasma cells <strong>of</strong> <strong>the</strong> nasal mucosa,which are avidly bound to <strong>the</strong> Fc-epsilon receptors onmast cells. This triggers degranulation <strong>of</strong> mast cells <strong>and</strong>releases <strong>the</strong> inflammatory mediators <strong>of</strong> <strong>the</strong> type I hypersensitivityreaction, causing rhinorrhoea <strong>and</strong> nasal obstruction.Microscopically, <strong>the</strong> nasal mucosa shows numerouseosinophils, abundant plasma <strong>and</strong> in some casesan increased number <strong>of</strong> mast cells. There is goblet cell


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 41hyperplasia <strong>of</strong> <strong>the</strong> respiratory epi<strong>the</strong>lium <strong>and</strong> <strong>the</strong> basementmembrane, which is destroyed in <strong>the</strong> acute phase,appears considerably thickened in <strong>the</strong> chronic phase.2.2.4 Atrophic RhinitisAtrophic rhinitis is a chronic inflammation <strong>of</strong> <strong>the</strong> nasalmucosa <strong>of</strong> unknown aetiology characterised by progressivenasal mucosal atrophy <strong>and</strong> by a thick, dense secretion,with a foetid smell <strong>and</strong> crusting [178]. Multiplefactors may be involved in <strong>the</strong> pathogenesis, includingchronic bacterial infections <strong>and</strong> nutritional deficiencies.Its incidence has markedly decreased in <strong>the</strong> last century,<strong>and</strong> nowadays most cases are secondary to trauma,surgery, granulomatous diseases, infection <strong>and</strong> radiationexposure [178]. Histologically, <strong>the</strong>re is non-specificchronic inflammatory infiltrate, squamous metaplasia<strong>of</strong> <strong>the</strong> surface epi<strong>the</strong>lium <strong>and</strong> <strong>of</strong> gl<strong>and</strong>ular excretoryducts, <strong>and</strong> atrophy <strong>of</strong> mucoserous gl<strong>and</strong>s [1, 69].2.2.5 Hypertrophic RhinitisThis term is applied to a condition <strong>of</strong> unknown aetiology,characterised by thickening <strong>of</strong> <strong>the</strong> sinonasal mucosaresulting from chronic inflammatory diseases [28, 71].Frequently, <strong>the</strong>se patients have undergone several sinusoperations, each time with limited success <strong>and</strong> subsequentrecurrence. Recurrent nasal polyposis is <strong>of</strong>ten associated.2.2.6 Non-Suppurative Chronic SinusitisChronic sinusitis is a complex, multifactorial disorderresulting from persistent acute inflammation or repeatedepisodes <strong>of</strong> acute or subacute sinusitis. There are usuallypredisposing factors like small sinus ostia, repeatedepisodes <strong>of</strong> common cold, allergy or acute sinusitisdetermining obstruction <strong>of</strong> <strong>the</strong> sinus ostia, reduction<strong>of</strong> ciliary activity (immotile cilia syndrome) <strong>and</strong> cysticfibrosis. The mucosal changes observed are variable<strong>and</strong> include basement membrane thickening, goblet cellhyperplasia, oedema <strong>of</strong> varying extent, inflammation(mostly lymphocytes <strong>and</strong> plasma cells) <strong>and</strong> polypoidchange <strong>of</strong> <strong>the</strong> mucosa [242].Allergic sinonasal polyps consist largely <strong>of</strong> myxoidoedematous tissue with pseudocysts containing eosinophilicproteinaceous fluid <strong>and</strong> infiltrates <strong>of</strong> inflammatorycells [115]. They are covered by respiratoryepi<strong>the</strong>lium with variable ulceration, goblet cellhyperplasia, squamous metaplasia <strong>and</strong> thickening <strong>of</strong><strong>the</strong> basement membranes. Seromucous gl<strong>and</strong>s <strong>and</strong>mucin-containing cysts may also occur. They arisemost frequently in <strong>the</strong> ethmoidal region <strong>and</strong> <strong>the</strong> upperpart <strong>of</strong> <strong>the</strong> nasal cavity. Allergic polyps usually exhibi<strong>the</strong>avy infiltration by eosinophils (Fig. 2.1a), markedthickening <strong>of</strong> <strong>the</strong> basement membranes <strong>and</strong> gobletcell hyperplasia. Most sinonasal polyps are <strong>of</strong> allergicorigin. Epi<strong>the</strong>lial dysplasia is present in a few cases.Granulomas may be present in polyps treated withintranasal injection, application <strong>of</strong> steroids or o<strong>the</strong>roily medications. Atypical fibroblasts with abundantcytoplasm, poorly defined cell borders <strong>and</strong> large pleomorphicnuclei are present in a small proportion <strong>of</strong>cases [183]. These atypical cells occur individually<strong>and</strong> are more frequently found close to blood vessels(Fig. 2.1b) or near <strong>the</strong> epi<strong>the</strong>lial surface. Such stromalatypia is a reactive phenomenon <strong>and</strong> it should not beconfused with sarcoma.2.3.2 Polyposis in MucoviscidosisNasal polyps in mucoviscidosis show cystic gl<strong>and</strong>s filledwith inspissated mucoid material <strong>and</strong> thickening <strong>of</strong> <strong>the</strong>basement membranes that surround <strong>the</strong> gl<strong>and</strong>s [22,189]. Some o<strong>the</strong>r polyps are <strong>of</strong> infective or chemical aetiology.The histological appearances <strong>of</strong> nasal polyps donot always correlate well with <strong>the</strong>ir aetiology.2.3.3 Polyposisin Immotile Cilia Syndrome<strong>and</strong> Kartagener’s SyndromeImmobile cilia syndrome (or primary ciliary dyskinesia)is a genetic disease affecting ciliary movement<strong>and</strong> resulting in respiratory infections <strong>and</strong> male infertility.Situs inversus may be associated ( Kartagener’ssyndrome). About 15% <strong>of</strong> patients develop nasal polypshistologically indistinguishable from o<strong>the</strong>r nasalpolyps. Ultrastructural analysis <strong>of</strong> nasal biopsies isneeded to identify <strong>the</strong> alterations in <strong>the</strong> architecture <strong>of</strong><strong>the</strong> cilium [177].2.3 Sinonasal Polyps2.3.1 Allergic Polyposis2.3.4 Antrochoanal PolypsAntrochoanal polyps are polyps that arise in <strong>the</strong>maxillary antrum <strong>and</strong> extend into <strong>the</strong> middle meatusprojecting posteriorly through <strong>the</strong> ipsilateral choana[106]. Antrochoanal polyps typically have prominentfibrous stroma surrounding thick-walled blood vessels


42 A. Cardesa · L. Alos · A. Franchi2aabFig. 2.1. a Allergic polyp with marked oedema <strong>of</strong> <strong>the</strong> stroma <strong>and</strong>heavy infiltration by eosinophils. b Atypical fibroblasts in an inflammatoryallergic polyp: enlarged fibroblasts with bizarre nuclei<strong>and</strong> occasional prominent nucleoli appear interspersed in granulationtissuebFig. 2.2 a Respiratory epi<strong>the</strong>lial adenomatoid hamartoma: gl<strong>and</strong>ular-likespaces lined by respiratory epi<strong>the</strong>lium <strong>and</strong> supportedby fibrous stroma. b Gl<strong>and</strong>ular hamartoma: abundant nodular aggregates<strong>of</strong> modified seromucous gl<strong>and</strong>s supported by slightly oedematousstroma[7]. In addition, scattered, enlarged, stromal cells withhyperchromatic nuclei are not an uncommon findingin this type <strong>of</strong> polyp [235]. Those polyps that arisein <strong>the</strong> maxillary antrum <strong>and</strong> extend into <strong>the</strong> middlemeatus projecting anteriorly are known as antronasalpolyps.2.4 Sinonasal Hamartomatous<strong>and</strong> Teratoid Lesions2.4.1 HamartomasSinonasal hamartomas are benign polypoid lesions inwhich well-developed branching gl<strong>and</strong>s <strong>and</strong>/or stromawith variable participation <strong>of</strong> different mesenchymalcomponents are present [266]. These lesions may resultfrom an exuberant hyperplastic reaction within <strong>the</strong>context <strong>of</strong> an inflammatory polyp. When <strong>the</strong> gl<strong>and</strong>s aremainly covered by ciliated respiratory epi<strong>the</strong>lium <strong>the</strong> lesionis termed “ respiratory epi<strong>the</strong>lial adenomatoid hamartoma”(Fig. 2.2a). If <strong>the</strong> gl<strong>and</strong>ular component consists<strong>of</strong> seromucous gl<strong>and</strong>s <strong>the</strong>y are known as “gl<strong>and</strong>ularhamartomas” (Fig. 2.2b). “Mesenchymal hamartomas”show predominance <strong>of</strong> skeletal muscle or <strong>of</strong> o<strong>the</strong>r mesenchymalelements.2.4.2 Teratoid LesionsDermoid cysts <strong>of</strong> <strong>the</strong> nose constitute 5.5–12% <strong>of</strong> those<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck region. More than half are detectedin children 6 years old or less, <strong>and</strong> approximately athird are present at birth. They occur most commonlyin <strong>the</strong> bridge <strong>of</strong> <strong>the</strong> nose <strong>and</strong> always in <strong>the</strong> midline [33,63, 88, 254, 278]. Dermoid cysts are lined with maturekeratinising squamous epi<strong>the</strong>lium <strong>and</strong> contain appendages<strong>of</strong> <strong>the</strong> skin in <strong>the</strong> cyst wall, but no endoderm.The lumen is filled with cheesy, yellow-white material.


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 43This lesion is differentiated from <strong>the</strong> very rare sinonasalteratoma by <strong>the</strong> limited variety <strong>of</strong> tissue types <strong>and</strong><strong>the</strong> absence <strong>of</strong> endodermal components [100]. Epidermalinclusion cysts do not contain adnexa. Dermoidcysts <strong>of</strong> <strong>the</strong> nose should also be distinguished from encephalocele.We are unaware <strong>of</strong> hairy polyps occurringin <strong>the</strong> nose.2.5 Pseudotumours2.5.1 MucoceleMucocele is a cyst filled with mucus that develops withina sinus cavity as <strong>the</strong> result <strong>of</strong> occlusion <strong>of</strong> <strong>the</strong> ostium.Most commonly it is due to infection, but may also resultfrom trauma or be congenital [109]. Retained secretionscause expansion <strong>of</strong> <strong>the</strong> sinus <strong>and</strong> bone erosion. Themost common sites <strong>of</strong> occurrence are <strong>the</strong> frontal <strong>and</strong> <strong>the</strong>sphenoidal sinuses. The cyst is lined by respiratory epi<strong>the</strong>liumthat shows prominent goblet-cell hyperplasia[158, 184]. Expansion <strong>of</strong> <strong>the</strong> cyst may cause atrophy <strong>and</strong>metaplasia <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium.2.5.2 Organising HaematomaOrganising haematoma, also known as “ cholesterolgranuloma” or “rhinitis caseosa”, is in most cases <strong>the</strong> result<strong>of</strong> occult submucosal haemorrhage in <strong>the</strong> maxillarysinus due to external trauma or tooth extraction [147].Resolution <strong>of</strong> <strong>the</strong> haematoma produces <strong>the</strong> formation <strong>of</strong>cholesterol granulomas <strong>and</strong> fibrosis, simulating a foreignbody reaction.2.5.4 MyospherulosisMyospherulosis is characterised by <strong>the</strong> presence <strong>of</strong> cystlikespaces lined by flattened histiocytes <strong>and</strong> containingclusters <strong>of</strong> brownish spherules resembling fungi [198,217, 230]. They lie loosely or within sacs formed by thinrefractile membranes. The brownish spherules do notstain with PAS or Gomori methanamine silver <strong>and</strong> <strong>the</strong>irmorphology does not correspond to any known fungus[228]. They are found within fibrous granulation tissue,which may show a foreign body reaction. The lesion isusually found in patients who have had previous operations[145]. It is now recognised that <strong>the</strong> spherules areextravasated red cells that have been altered by interactionwith traumatised fat or petrolatum-based ointments<strong>and</strong> gauzes used in surgical procedures.2.5.5 Eosinophilic Angiocentric FibrosisEosinophilic angiocentric fibrosis is a rare, chronic, benign,idiopathic condition <strong>of</strong> <strong>the</strong> upper respiratory tractoccurring predominantly in adult women [214, 248].Initially, <strong>the</strong> histologic picture is characterised by nonnecrotisingeosinophilic vasculitis involving capillaries<strong>and</strong> venules <strong>of</strong> <strong>the</strong> sinonasal mucosa, accompanied by aninflammatory infiltrate with lymphocytes, plasma cells,histiocytes <strong>and</strong> occasional neutrophils [214]. In late lesions<strong>the</strong>re is a characteristic obliterative perivascularonion-skin fibrosis, while <strong>the</strong> inflammatory infiltrate isless dense <strong>and</strong> eosinophils predominate [214]. The differentialdiagnosis includes reactive processes <strong>of</strong> <strong>the</strong> sinonasalmucosa, like Wegener’s granulomatosis, Churg-Strauss syndrome, Kimura disease, <strong>and</strong> angiolymphoidhyperplasia with eosinophilia.2.5.3 AmyloidosisIsolated amyloid deposition in <strong>the</strong> sinonasal mucosa isa rare event, with about 20 cases reported in <strong>the</strong> English-languageliterature [180, 258]. Grossly, <strong>the</strong> lesionappears as a friable tumour-like mass, with a tendencyto bleed. Histologically, <strong>the</strong>re is a deposition <strong>of</strong> intenselyeosinophilic material in <strong>the</strong> stroma, around blood vessels<strong>and</strong> around ducts <strong>of</strong> <strong>the</strong> mucoserous gl<strong>and</strong>s, whichis <strong>of</strong>ten associated with diffuse chronic inflammation<strong>and</strong> foreign body granulomatous reaction. Amyloidstains orange with Congo red, <strong>and</strong> showed apple greenbirefringence under polarised light examination. Immunohistochemistrymay help to identify <strong>the</strong> type <strong>of</strong>amyloid deposition.2.5.6 Heterotopic Brain TissueThis lesion mostly occurs in young children, usually <strong>the</strong>result <strong>of</strong> a congenital abnormality related to a variant <strong>of</strong>meningoencephalocele [136, 196]. Commonly used synonymsare glial heterotopia <strong>and</strong> nasal glioma, although<strong>the</strong> latter is a misnomer. The lesion mainly arises at <strong>the</strong>base <strong>of</strong> <strong>the</strong> nose or in <strong>the</strong> upper part <strong>of</strong> <strong>the</strong> nasal cavity,<strong>and</strong> grossly may be polypoid. Histologically, it is mostlycomposed <strong>of</strong> a mixture <strong>of</strong> astrocytes, glial fibres <strong>and</strong>fibrous connective tissue. Multinucleated glial cells arefrequently found. Some glial cells can have large nucleiresembling nerve cells. Immunostaining for glial fibrillaryacidic protein is a helpful diagnostic adjunct. A fewtrue nerve cells or even ependymal elements can rarelybe identified. Mitoses are not found.


44 A. Cardesa · L. Alos · A. Franchi2aPAS or Gomori methanamine silver <strong>the</strong> fungi appearas dichotomously branching septate hyphae 6–8 mwide. Aspergillosis may occur as a non-invasive diseasein which a mass <strong>of</strong> fungal hyphae (fungal ball) is presentin a sinus (Fig. 2.3a). Invasive aspergillosis is seenmore <strong>of</strong>ten in immunocompromised patients, associatedwith destructive inflammation <strong>of</strong> <strong>the</strong> sinonasaltissues [223]. The disease may also occur as an allergicmucinous sinusitis in which <strong>the</strong> sinuses contain masses<strong>of</strong> inspissated mucus with abundant eosinophils, Charcot-Leydencrystals (Fig. 2.3b), necrotic cell debris <strong>and</strong>scarce fungal hyphae (Fig. 2.3c) [137, 172]. The sinusmucosa shows inflammatory changes without fungalinvasion.b2.6.2 MucormycosisMucormycosis is caused by fungi <strong>of</strong> <strong>the</strong> class Zygomycetes<strong>and</strong> order Mucorales [73]. The most common speciescausing sinonasal infection are Rhizopus arrhizus<strong>and</strong> Rhizopus oryzae. In sections stained with PAS orGomori methanamine silver <strong>the</strong> fungi are seen as nonseptatehyphae measuring 10- to 20-m wide, usuallybranching at right angles. Infection is usually opportunistic<strong>and</strong> causes rapidly progressive disease in poorlycontrolled diabetics <strong>and</strong> immunocompromised patients.The fungus has a tendency to invade blood vessels causingthrombosis; <strong>the</strong> affected tissues may exhibit coagulativenecrosis <strong>and</strong> haemorrhage.cFig. 2.3. a Sinonasal aspergilloma: densely packed branching hyphae<strong>of</strong> aspergillus forming a fungal ball. (Gomori’s me<strong>the</strong>naminesilver). b Sinonasal allergic mucinosis: dense aggregates <strong>of</strong> eosinophilicleukocytes distributed between pools <strong>of</strong> mucin. At <strong>the</strong> centre,one Charcot-Leyden crystal. c Allergic fungal sinusitis: scarcefungal hyphae found after diligent search in a lake <strong>of</strong> mucin (Gomori’sme<strong>the</strong>namine silver)2.6 Fungal Diseases2.6.1 AspergillosisAspergillosis is caused by Aspergillus fumigatus, Aspergillusniger <strong>and</strong> o<strong>the</strong>r species. In sections stained with2.6.3 RhinosporidiosisRhinosporidiosis is caused by <strong>the</strong> endosporulatingfungus Rhinosporidium seeberi. The lesions are polypoid<strong>and</strong> occur principally in <strong>the</strong> nasal cavity [21, 161].They are characterised by <strong>the</strong> presence <strong>of</strong> thick-walledsporangia measuring 50–350 m in diameter <strong>and</strong> containingnumerous mucicarminophilic spores. They areassociated with a heavy chronic inflammatory reactionwith occasional foci <strong>of</strong> suppuration <strong>and</strong> foreign body giantcell reaction.2.7 HIV-Related InfectionsSinonasal infections are frequently observed in HIV patients,are <strong>of</strong>ten asymptomatic <strong>and</strong> tend to be recurrentor refractory [281]. They are due to various pathogensincluding cytomegalovirus [164], Staphylococcus aureus,fungi (Aspergillus) [170] <strong>and</strong> parasites (Microsporidium,Cryptosporidium) [66].


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 452.8 Mid-Facial NecrotisingGranulomatous LesionsTuberculosis <strong>of</strong> head <strong>and</strong> neck occurs infrequently <strong>and</strong>involvement <strong>of</strong> <strong>the</strong> nose is rare, representing in mostcases a secondary event to pulmonary involvement[231]. In most cases <strong>the</strong>re is a polyp <strong>of</strong> <strong>the</strong> nasal septumor an ulcerated granular lesion. Presence <strong>of</strong> intracranialextension may lead to a clinical diagnosis <strong>of</strong> malignancy[19]. Microscopically, <strong>the</strong>re are caseating giant cellgranulomas in which acid-fast bacilli may occasionallybe identified. The definitive diagnosis is made by isolatingMycobacterium tuberculosis from tissue removedduring biopsy.2.8.1 Wegener’s GranulomatosisWegener’s granulomatosis is an immunologically mediatedinflammatory disease characterised by granulomatousvasculitis <strong>of</strong> <strong>the</strong> upper <strong>and</strong> lower respiratorytracts toge<strong>the</strong>r with glomerulonephritis. Variable degrees<strong>of</strong> disseminated vasculitis involving both smallarteries <strong>and</strong> veins may also occur. The lesions in <strong>the</strong>upper respiratory tract are ulcerative <strong>and</strong> destructive<strong>and</strong> occur mainly in <strong>the</strong> nasal cavity <strong>and</strong> paranasalsinuses. The hallmarks <strong>of</strong> Wegener’s granulomatosisare <strong>the</strong> presence <strong>of</strong> geographic necrosis surrounded bypalisaded histiocytes, granulomas <strong>and</strong> scattered giantcells, vasculitis with fibrinoid necrosis or infiltration<strong>of</strong> vessel walls by inflammatory cells, neutrophilic microabscesses<strong>and</strong> a mixed inflammatory infiltrate withvariable fibrosis [57, 165]. Stains for acid fast bacilli <strong>and</strong>fungi are negative. There is no cytological atypia. Theclassic histological features <strong>of</strong> Wegener granulomatosisare not present in many biopsy specimens. Repeatbiopsies <strong>and</strong> clinical correlations are <strong>of</strong>ten essential forearly diagnosis. The disease may be restricted to <strong>the</strong>upper respiratory tract in <strong>the</strong> early stages. A high percentage<strong>of</strong> patients develop c-ANCA. More details areto be found in Chap. 3.2.8.2 Lepromatous LeprosyLepromatous leprosy is <strong>the</strong> most frequent form <strong>of</strong> thistype <strong>of</strong> disease involving <strong>the</strong> nasal cavity [101]. It ischaracterised by nodular masses <strong>of</strong> foamy macrophages(lepra cells) in which large numbers <strong>of</strong> acid fast bacilli(Mycobacterium leprae) are demonstrable by <strong>the</strong> modifiedZiehl-Neelsen method. Tuberculoid leprosy is characterisedby non-caseating granulomas <strong>and</strong> <strong>the</strong> indeterminatevariant by a non-specific chronic inflammatoryreaction; acid fast bacilli are seldom demonstrable in<strong>the</strong>se types.2.8.3 Tuberculosis2.8.4 SarcoidosisSarcoidosis is a chronic multisystem granulomatousdisorder that has a predilection for pulmonary <strong>and</strong> upperrespiratory tract mucosa. The sinonasal mucosais rarely involved, <strong>and</strong> most patients have generaliseddisease [143]. Discrete non-caseating granulomatacomposed predominantly <strong>of</strong> epi<strong>the</strong>lioid histiocyteswith multinucleated giant cells <strong>and</strong> a peripheral rim <strong>of</strong>lymphocytes are present in <strong>the</strong> mucosa. Stains for acidfastbacilli are negative. The differential diagnosis includeso<strong>the</strong>r granulomatous disorders, like tuberculosis,leprosy, Wegener’s granulomatosis <strong>and</strong> cholesterolgranuloma [57].2.8.5 RhinoscleromaRhinoscleroma is caused by Klebsiella rhinoscleromatis[21], a capsulated gram-negative bacillus. Largenodular tumour-like masses are found in <strong>the</strong> nasalcavity <strong>and</strong> less <strong>of</strong>ten in o<strong>the</strong>r parts <strong>of</strong> <strong>the</strong> upper respiratorytract. They contain large macrophages withabundant clear or vacuolated cytoplasm (Mikuliczcells). The causative organism may be identified by<strong>the</strong> Warthin-Starry staining method or by immunostainingfor <strong>the</strong> Klebsiella capsular antigen. Thereis heavy infiltration by chronic inflammatory cells,mainly plasma cells showing numerous Russell bodies.2.8.6 LeishmaniasisLeishmaniasis <strong>of</strong> <strong>the</strong> nasal region, when seen in Mediterraneancountries, is mostly in <strong>the</strong> form <strong>of</strong> an “orientalsore” caused by Leishmania tropica. In Central <strong>and</strong> SouthAmerica it is mostly seen in <strong>the</strong> form <strong>of</strong> mucocutaneousleishmaniasis caused by Leishmania braziliensis [153,197]. The protozoan parasite is seen in <strong>the</strong> cytoplasm<strong>of</strong> histiocytes or extracellularly, measures 1.5–3.0 m inits maximum dimension <strong>and</strong> has a nucleus <strong>and</strong> a rodshapedkinetoplast that stains positively with Giemsa.The kinetoplast is more readily identified in Giemsastainedsmears <strong>of</strong> exudates or scrapings than in paraffinsections. The lesions, commonly found in <strong>the</strong> nasal mucosa<strong>and</strong> facial skin, are associated with chronic inflammatoryreaction <strong>and</strong> granuloma formation. They are ingeneral circumscribed <strong>and</strong> self-involutive in <strong>the</strong> case <strong>of</strong><strong>the</strong> “oriental sore” <strong>and</strong> markedly destructive in mucocutaneousleishmaniasis.


46 A. Cardesa · L. Alos · A. Franchi22.8.7 Cocaine AbuseCocaine abuse may be associated with severe nasalnecrotising inflammation [225]. Endoscopically, <strong>the</strong>re isatrophy <strong>of</strong> <strong>the</strong> inferior <strong>and</strong> middle turbinates <strong>and</strong> ulceration<strong>of</strong> <strong>the</strong> nasal septum. Histologically, areas <strong>of</strong> acute<strong>and</strong> chronic inflammation are found; however, vasculitisis minimal or absent <strong>and</strong> granulomas may be present.The lesion may be confused with Wegener’s granulomatosis.2.8.8 Local Steroid InjectionsA granulomatous lesion <strong>of</strong> <strong>the</strong> nasal mucous membraneshas been observed in patients treated with injections <strong>of</strong>steroid preparations [272]. There is a central deposition <strong>of</strong>amorphous material bordered by histiocytes <strong>and</strong> foreignbody giant cells. Occasional particles <strong>of</strong> birefringent crystallinematerial may be present. Special stains should beperformed to exclude <strong>the</strong> presence <strong>of</strong> micro-organisms.2.9 Benign Epi<strong>the</strong>lial Neoplasms2.9.1 Sinonasal PapillomasSinonasal papillomas may be divided into squamous cellpapillomas <strong>of</strong> <strong>the</strong> nasal vestibule <strong>and</strong> schneiderian papillomas<strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> paranasal sinuses [121].The first are covered by epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong> skin surface.The latter are lined by well-differentiated respiratoryepi<strong>the</strong>lium (referred to as <strong>the</strong> schneiderian membrane)<strong>and</strong> comprise three histopathological types: exophytic,inverted <strong>and</strong> oncocytic. The histopathological featuresthat clearly differentiate between <strong>the</strong> three types <strong>of</strong> schneiderianpapillomas have been well documented [173].Human papilloma virus (HPV) types 6 <strong>and</strong> 11 are involvedin <strong>the</strong> pathogenesis <strong>of</strong> exophytic papillomas, butnot in <strong>the</strong> o<strong>the</strong>r two variants <strong>of</strong> schneiderian papillomas[35, 89, 128]. All oncocytic papillomas examined havebeen HPV-negative [35, 128, 221].2.9.1.1 Squamous Cell PapillomaICD-O:8052/0Squamous cell papillomas are located in <strong>the</strong> nasal vestibule<strong>and</strong> are formed by keratinising stratified squamousepi<strong>the</strong>lium <strong>of</strong> <strong>the</strong> skin surface [122]. They are exophytic<strong>and</strong> consist <strong>of</strong> a thickened layer <strong>of</strong> differentiated squamousepi<strong>the</strong>lium, without evidence <strong>of</strong> atypia or mitoses,which is supported by arborescent stalks <strong>of</strong> fibrovascularstroma. Varying degrees <strong>of</strong> keratinisation are present<strong>and</strong> ei<strong>the</strong>r hyperkeratosis, parakeratosis or both may beseen. They are benign, rarely recur after simple excision.Its main differential diagnosis is <strong>the</strong> exophytic schneiderianpapilloma.2.9.1.2 Exophytic PapillomaICD-O:8121/0Exophytic papilloma, also known as “everted” or “fungiform”papilloma, is a single, warty tumour measuringup to 1.5 cm in diameter, arising most frequentlyat <strong>the</strong> nasal septum <strong>and</strong> only very rarely in <strong>the</strong> lateralnasal walls or in paranasal sinuses [122]. Males are predominantlyaffected. Patients tend to be younger thanwith o<strong>the</strong>r types <strong>of</strong> schneiderian papilloma. Exophyticpapillomas are almost always unilateral [54]. No side ispreferred <strong>and</strong> bilaterality is exceptional. The tumour iscomposed <strong>of</strong> branching papillary structures, with papillaecovered by stratified non-keratinising squamousepi<strong>the</strong>lium, admixed with intermediate or transitionalcells <strong>and</strong> with ciliated respiratory epi<strong>the</strong>lium that containsinterspersed mucin-secreting cells. Koilocytosisis not infrequently found in <strong>the</strong> squamous epi<strong>the</strong>lium.Seromucinous gl<strong>and</strong>s are abundantly found when <strong>the</strong>underlying submucosa is removed.The two main differential diagnoses are invertedpapilloma <strong>and</strong> oncocytic papilloma. Nei<strong>the</strong>r <strong>the</strong> invaginatedpattern <strong>of</strong> growth <strong>of</strong> inverted papillomas nor <strong>the</strong>oncocytic columnar epi<strong>the</strong>lium <strong>of</strong> oncocytic papillomasare found in exophytic papillomas [173]. Cylindrical cellcarcinoma can be easily ruled out by <strong>the</strong> lack <strong>of</strong> atypia<strong>and</strong> invasion. Wide surgical excision is <strong>the</strong> best choice<strong>of</strong> treatment to avoid recurrences. Recurrences occur inabout 20–40% <strong>of</strong> cases, which is less than in invertedpapillomas. Malignant transformation almost never occursin exophytic papillomas.2.9.1.3 Inverted PapillomaICD-O:8121/0Inverted papilloma is <strong>the</strong> most common type <strong>of</strong> schneiderianpapilloma. This lesion occurs almost exclusivelyin <strong>the</strong> lateral wall <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> in <strong>the</strong> paranasalsinuses, although on rare occasions it may alsoarise on <strong>the</strong> nasal septum [226]. Grossly, <strong>the</strong>y frequentlyhave a polypoid appearance, but <strong>the</strong>y differ from nasalpolyps <strong>of</strong> <strong>the</strong> common type by <strong>the</strong>ir histological features.Inverted papillomas are composed <strong>of</strong> invaginatingcrypts, cords <strong>and</strong> nests covered by non-keratinisingsquamous epi<strong>the</strong>lium, which alternates with columnarciliated respiratory epi<strong>the</strong>lium <strong>and</strong> with intermediate ortransitional epi<strong>the</strong>lium (Fig. 2.4a). This newly formedduct system is similar to <strong>the</strong> embryonic development<strong>of</strong> <strong>the</strong> nasal mucosa [240]. The multilayered epi<strong>the</strong>liumtypically contains mucous cells <strong>and</strong> mucin-filled micro-


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 47cysts. The invagination <strong>of</strong> <strong>the</strong> mucosa may result in <strong>the</strong>presence <strong>of</strong> apparently discontinuous cell masses lyingdeep to <strong>the</strong> epi<strong>the</strong>lial surface, but <strong>the</strong> basement membraneis intact <strong>and</strong> may be shown in continuity with that<strong>of</strong> <strong>the</strong> surface epi<strong>the</strong>lium [226]. An inverted growth is<strong>the</strong> hallmark <strong>of</strong> inverted papilloma, but varying degrees<strong>of</strong> papillary growth may be seen at <strong>the</strong> surface [54]. Thesurface is characteristically lined by a respiratory type<strong>of</strong> epi<strong>the</strong>lium; never<strong>the</strong>less, foci <strong>of</strong> surface keratinisationare occasionally present [122]. A few regular mitosesmay be found in <strong>the</strong> basal <strong>and</strong> parabasal layers. Although<strong>the</strong> nuclei may show mild nuclear irregularities<strong>and</strong> hyperchromatism, no disturbances <strong>of</strong> <strong>the</strong> cellularpolarity are found. An abundant <strong>and</strong> oedematous connectivetissue stroma is a common feature <strong>of</strong> invertedpapillomas. It usually contains macrophages <strong>and</strong> neutrophils,but eosinophils may also be present. This inflammatoryinfiltrate may also be present between <strong>the</strong>epi<strong>the</strong>lial cells, within <strong>the</strong> dilated lumens <strong>of</strong> invaginatedcrypts, <strong>and</strong> within <strong>the</strong> numerous microcysts that usuallyoccur in <strong>the</strong> respiratory epi<strong>the</strong>lium. Seromucinousgl<strong>and</strong>s are absent, but branching gl<strong>and</strong> ducts are <strong>of</strong>tenpresent. The tumour grows by extension to involve <strong>the</strong>contiguous sinonasal epi<strong>the</strong>lium.If treated only by local surgical excision, recurrenceoccurs in up 75% <strong>of</strong> cases. Therefore, lateral rhinotomy<strong>and</strong> medial maxillectomy are advisable for tumours <strong>of</strong><strong>the</strong> lateral nasal wall [236]. Carcinoma develops in about10–15% <strong>of</strong> inverted papillomas [122, 211, 236]. Carcinomamay coexist with inverted papilloma at <strong>the</strong> initialpresentation or originate subsequently [122, 273].According to <strong>the</strong> experience <strong>of</strong> Michaels <strong>and</strong> Hellquist[171], carcinoma does not usually develop in <strong>the</strong> course<strong>of</strong> recurrences <strong>of</strong> inverted papilloma. In <strong>the</strong> presence<strong>of</strong> severe atypia or marked keratinisation in an invertedpapilloma malignant transformation is always suspected(Fig. 2.4b). In <strong>the</strong>se instances <strong>the</strong> entire specimenshould be thoroughly examined to exclude an associatedcarcinoma. Most associated carcinomas are squamous[204], although o<strong>the</strong>r types may also occur such asverrucous carcinoma [192].2.9.1.4 Oncocytic PapillomaabFig. 2.4. a Inverted papilloma: prominent invaginating cryptslined by hyperplastic respiratory epi<strong>the</strong>lium are supported by oedematousconnective tissue. b Squamous cell carcinoma ex invertedpapilloma: remnants <strong>of</strong> benign invaginating crypts are seen betweeninvasive cords <strong>of</strong> squamous epi<strong>the</strong>liumICD-O:8121/1Oncocytic papilloma, also known as “columnar” or “cylindric”cell papilloma [226], is <strong>the</strong> least common type <strong>of</strong>schneiderian papilloma. It constitutes less than 5% <strong>of</strong> allsinonasal papillomas [18, 122, 173, 262]. Both sexes areequally affected. Bilaterality has not been documented.Tumours are in general small, although occasionally mayreach various centimetre measurements in <strong>the</strong>ir greatestdimension. They are composed <strong>of</strong> exophytic fronds<strong>and</strong> endophytic invaginations lined by pseudostratifiedor multilayered columnar cells with prominent oncocyticfeatures. The cells have uniform hyperchromaticnuclei <strong>and</strong> abundant eosinophilic, occasionally granular,cytoplasm that contains abundant mitochondria<strong>and</strong> stains for <strong>the</strong> mitochondrial enzyme cytochromeC oxidase [59]. Goblet cells are not found. Cilia may beoccasionally encountered on <strong>the</strong> superficial epi<strong>the</strong>liallayer. Intraepi<strong>the</strong>lial microcysts containing mucin <strong>and</strong>neutrophils are usually present. These microcysts arelarger than <strong>the</strong> similar structures also seen in invertedpapilloma. The tumour resembles inverted papilloma inits sites <strong>of</strong> occurrence, <strong>the</strong> lateral wall <strong>of</strong> <strong>the</strong> nasal cavity<strong>and</strong> <strong>the</strong> maxillary antrum. The rate <strong>of</strong> recurrence isconsidered to be 36%, which is lower than in invertedpapilloma. The low frequency <strong>of</strong> this tumour makes itdifficult to evaluate its true malignant potential, whichseems to be similar to that <strong>of</strong> inverted papilloma [262].Atypical hyperplasia <strong>and</strong> carcinoma in situ changescan be occasionally found (Fig. 2.5). Surgical excisionwith wide margins is <strong>the</strong> treatment <strong>of</strong> choice. Invasivesquamous cell carcinoma, high-grade mucoepidermoid


48 A. Cardesa · L. Alos · A. Franchi2carcinoma <strong>and</strong> undifferentiated carcinoma have beenreported in association with oncocytic papilloma [18,122, 135, 265, 274].2.9.2 Salivary-Type AdenomasPleomorphic adenoma is <strong>the</strong> most frequent benign gl<strong>and</strong>ulartumour <strong>of</strong> <strong>the</strong> sinonasal region. Most arise on <strong>the</strong>nasal septum <strong>and</strong> <strong>the</strong> rest on <strong>the</strong> lateral nasal wall orturbinates. Origin in <strong>the</strong> maxillary antrum is rare. Therecurrence rate <strong>of</strong> sinonasal pleomorphic adenoma ismuch lower than for its counterpart in <strong>the</strong> major salivarygl<strong>and</strong>s [56, 109]. Rare examples <strong>of</strong> sinonasal oncocytoma,myoepi<strong>the</strong>lioma <strong>and</strong> basal cell adenoma havebeen reported [27, 55, 103, 277], as well as one case <strong>of</strong>sinonasal myoepi<strong>the</strong>lioma transformed into myoepi<strong>the</strong>lialcarcinoma after various recurrences [9].2.9.3 Pituitary AdenomasThe rare pituitary adenomas <strong>of</strong> <strong>the</strong> sinonasal region arein most instances extensions from intrasellar tumours.Very unusually, <strong>the</strong>y arise from ectopic pituitary tissueas tumours from <strong>the</strong> sphenoid sinus or <strong>the</strong> nasal cavity.Histologically, <strong>the</strong>y are similar to tumours within <strong>the</strong>sella [61, 151].2.10 Benign SinonasalS<strong>of</strong>t Tissue Neoplasms2.10.1 HaemangiomasHaemangiomas <strong>of</strong> <strong>the</strong> upper respiratory tract may be <strong>of</strong><strong>the</strong> capillary, cavernous or venous types [90]. The mostcommon type is <strong>the</strong> capillary haemangioma whichconsists <strong>of</strong> lobules <strong>of</strong> blood-filled capillaries separatedby loose connective tissue. The lesion should be distinguishedfrom granulation tissue <strong>and</strong> from <strong>the</strong> vascularectasias found in Rendu-Weber-Osler disease.2.10.2 Haemangiopericytoma(Glomangiopericytoma)ICD-O:9150/1Haemangiopericytoma is characterised by <strong>the</strong> proliferation<strong>of</strong> oval, polyhedral or spindle-shaped cells enmeshedby collagen type IV fibres <strong>and</strong> arranged around vascularchannels that are lined by a single layer <strong>of</strong> endo<strong>the</strong>lialcells. The tumour contains numerous thin-walled bloodvessels <strong>and</strong> <strong>the</strong> tumour cells, typically arranged around<strong>the</strong> blood vessels, are <strong>of</strong> uniform size with regular ovalFig. 2.5. Oncocytic papilloma with atypical cells: papillary stalkscovered by columnar cells with frequent atypical nuclei <strong>and</strong> oncocyticcytoplasm-forming microcystsor elongated nuclei <strong>and</strong> pale cytoplasm (Fig. 2.6). Thecells may also be arranged in short, haphazard fasciclesor in sheets <strong>of</strong> closely packed cells containing compressedcapillaries. Areas <strong>of</strong> poor cellularity, myxoid change <strong>and</strong>fibrosis are not uncommon. The tumour cells are entirelysituated outside <strong>the</strong> capillaries, which are lined by a singlelayer or normal-looking endo<strong>the</strong>lium. This feature, wellshown by reticulin staining or by anti-collagen IV antibodies,helps to distinguish <strong>the</strong> tumour from angiosarcoma.The distinction from o<strong>the</strong>r well-vascularised mesenchymaltumours is usually made by exclusion. Haemangiopericytomas<strong>of</strong> <strong>the</strong> nasal cavity are generally lessaggressive than those occurring elsewhere. They exhibit amore orderly structure with minimal mitotic activity, buttend to recur after removal <strong>and</strong> may rarely metastasise[249]. Muscle-specific actin is focally positive in tumourcells. The term glomangiopericytoma has been recentlyproposed for this entity [267a].2.10.3 Solitary Fibrous TumourICD-O:8815/0Solitary fibrous tumour <strong>of</strong> <strong>the</strong> nose, paranasal sinuses<strong>and</strong> nasopharynx is in most instances a benign fibroblasticproliferation with variable cellularity <strong>and</strong> vascularity(Fig. 2.7) having features identical to those <strong>of</strong>solitary fibrous tumour <strong>of</strong> <strong>the</strong> pleura [8, 168, 279]. Itsmain differential diagnoses are sinonasal haemangiopericytoma<strong>and</strong> nasopharyngeal angi<strong>of</strong>ibroma.2.10.4 Desmoid FibromatosisICD-O:8821/1Desmoid fibromatoses are a group <strong>of</strong> non-metastasisingunencapsulated fibrous tissue proliferations that have a


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 49tendency towards local invasion <strong>and</strong> recurrence, whichrarely arise in <strong>the</strong> sinonasal mucosa [96]. They compriseinterlacing fascicles <strong>of</strong> bl<strong>and</strong> spindle-shaped fibroblasts,in a collagenous or myxoid background. The main differentialdiagnoses are fibrosarcoma <strong>and</strong> reactive fibrosis.Desmoid fibromatosis <strong>of</strong> <strong>the</strong> sinonasal tract showslower recurrence rates than desmoid fibromatoses arisingin o<strong>the</strong>r locations.2.10.5 Fibrous HistiocytomaICD-O:8830/0Benign fibrous histiocytoma presents as a yellow-tan noduleor polyp, most commonly causing nasal obstruction orbleeding [201]. It is composed <strong>of</strong> spindle-shaped cells producinga storiform pattern admixed with histiocytic cells<strong>and</strong> multinucleated giant cells. Distinction from o<strong>the</strong>r benignsinonasal spindle cell proliferations is largely basedon <strong>the</strong> immunohistochemical findings. Benign fibroushistiocytomas may recur if incompletely excised.Fig. 2.6. Haemangiopericytoma: interconnected thin-walledblood vessels surrounded by uniform spindle-shaped cells withoval or elongated nuclei2.10.6 LeiomyomaICD-O:8890/0Sinonasal leiomyoma is a rare tumour occurring inadults, preferentially involving <strong>the</strong> nasal cavities, withnon-specific symptoms <strong>of</strong> nasal obstruction [91]. Itsmorphologic <strong>and</strong> immunohistochemical pr<strong>of</strong>ile is identicalto that <strong>of</strong> leiomyomas <strong>of</strong> o<strong>the</strong>r sites. It has been postulatedthat <strong>the</strong>y may originate from blood vessel walls.Distinction from sinonasal leiomyosarcoma is basedon <strong>the</strong> absence <strong>of</strong> atypia <strong>and</strong> mitoses. Huang <strong>and</strong> Antonescuhave proposed separating a category <strong>of</strong> smoothmuscle tumours <strong>of</strong> uncertain malignant potential, characterisedby <strong>the</strong> presence <strong>of</strong> 1–4 mitotic figures/10 highpower fields, that tend to pursue a more aggressive behaviourthan leiomyomas [119].2.10.7 Schwannoma <strong>and</strong> Neur<strong>of</strong>ibromaICD-O:9560/0, 9540/0About 4% <strong>of</strong> schwannomas <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck regionarise in <strong>the</strong> sinonasal tract [202]. They usually presentas polypoid lesions involving <strong>the</strong> nasal cavity <strong>and</strong>/ora paranasal sinus, with non-specific symptoms <strong>of</strong> obstruction,compression, or extension in <strong>the</strong> surroundingstructures [202]. Histologically, <strong>the</strong> tumour is composed<strong>of</strong> elongated wavy-shaped monomorphic spindle cells,with eosinophilic cytoplasm <strong>and</strong> oval nucleus. Antonitype A <strong>and</strong> type B areas usually coexist within <strong>the</strong> lesion,<strong>and</strong> nuclear palisading may be present. Focal degenerativenuclear atypia has been described [108], whilemitotic activity is absent to low. A consistently reportedFig. 2.7. Solitary fibrous tumour: fibroblastic proliferation, collagenproduction <strong>and</strong> dilated blood vessels. Identical features to <strong>the</strong>pleural counterpartfeature <strong>of</strong> sinonasal schwannomas is <strong>the</strong> lack <strong>of</strong> tumourencapsulation that determines an apparently infiltrativegrowth pattern [36, 108]. Immunohistochemically,sinonasal schwannoma is intensely reactive for S-100protein [108]. The differential diagnosis includes o<strong>the</strong>rspindle cell lesions <strong>of</strong> <strong>the</strong> sinonasal mucosa, like juvenileangi<strong>of</strong>ibroma, solitary fibrous tumour <strong>and</strong> leiomyoma.Particular care should be taken when evaluating cellularschwannomas with a predominance <strong>of</strong> Antoni typeA areas, which should not be confused with malignantspindle cell neoplasms, like fibrosarcoma, leiomyosarcoma,malignant peripheral nerve sheath tumour, <strong>and</strong>spindle cell melanoma.Neur<strong>of</strong>ibromas <strong>of</strong> <strong>the</strong> sinonasal mucosa are usuallynot associated with <strong>the</strong> Von Recklinghausen syndrome,<strong>and</strong> appear as unencapsulated lesions composed <strong>of</strong> a mixture<strong>of</strong> Schwann cells <strong>and</strong> fibroblasts embedded in a predominantlymyxoid stroma [117, 202]. Due to <strong>the</strong> overlapping<strong>of</strong> <strong>the</strong> histologic features, it may be difficult to differ-


50 A. Cardesa · L. Alos · A. Franchisinuses. Histologically, <strong>the</strong>y are similar to paragangliomaselsewhere [12, 98, 187].22.10.10 Juvenile Angi<strong>of</strong>ibromaabFig. 2.8. Juvenile angi<strong>of</strong>ibroma. a Polypoid mass <strong>of</strong> white-redcut surface <strong>and</strong> rubbery consistency. b Vascular elements embeddedin fibrous tissue showing intravascular microembolisation, atreatment modality prior to surgeryentiate neur<strong>of</strong>ibromas from schwannomas <strong>of</strong> <strong>the</strong> sinonasalmucosa. Neur<strong>of</strong>ibromas should also be distinguishedfrom myxomas, which are S-100 protein-negative.2.10.8 MeningiomaICD-O:9530/0Meningiomas <strong>of</strong> <strong>the</strong> sinonasal tract may extend directlyfrom <strong>the</strong> central nervous system or arise from ectopicextracranial tissue. Although always rare, <strong>the</strong>y are morecommonly seen in <strong>the</strong> orbit, ear <strong>and</strong> skin <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck than in <strong>the</strong> sinonasal tract. Histologically, <strong>the</strong>y aresimilar to meningiomas elsewhere, <strong>the</strong> meningo<strong>the</strong>lialtype being <strong>the</strong> most frequent. Sinonasal meningiomastend to occur in younger patients than intracranial meningiomas[118, 203].2.10.9 ParagangliomaICD-O:8680/1There are few reports on nasal paragangliomas. The tumoursoriginate in <strong>the</strong> middle turbinates <strong>and</strong> ethmoidICD-O:9160/0Juvenile nasopharyngeal angi<strong>of</strong>ibroma arises in <strong>the</strong>confluence <strong>of</strong> <strong>the</strong> posterolateral nasal wall <strong>and</strong> <strong>the</strong>lateral nasopharynx <strong>and</strong> occurs almost exclusively inmales during adolescence [90, 125]. The tumour is sessileor polypoid (Fig. 2.8a) <strong>and</strong> is histologically benign,but has a tendency to recur <strong>and</strong> is locally destructive,causing pressure necrosis <strong>of</strong> adjacent s<strong>of</strong>t tissue <strong>and</strong>bone. It may occasionally extend into paranasal sinuses,orbit <strong>and</strong> cranial fossae. It is composed <strong>of</strong> vascular <strong>and</strong>fibrous elements in varying proportions. The vessels in<strong>the</strong> superficial portions <strong>of</strong> <strong>the</strong> tumour are mainly gapingcapillaries that may become compressed with increasingstromal fibrosis. Thick-walled vessels withoutelastic membranes <strong>and</strong> with irregular, incomplete orabsent muscle coats <strong>and</strong> focal intimal thickenings areusually present in <strong>the</strong> deeper portions <strong>of</strong> <strong>the</strong> tumour.These vessels resemble those normally seen in <strong>the</strong> submucosa<strong>of</strong> <strong>the</strong> nasal conchae. The vascular elements areembedded in fibrous tissue, which varies in cellularity<strong>and</strong> collagenisation. Stellate fibroblast-like cells are <strong>of</strong>tenpresent close to <strong>the</strong> blood vessels. The fibroblasticcells <strong>of</strong> nasopharyngeal angi<strong>of</strong>ibroma are stronglypositive for testosterone receptors [120]. Ultrastructurally,<strong>the</strong> nuclei <strong>of</strong> angi<strong>of</strong>ibroma contain characteristicdense granules [251]. Occasionally, <strong>the</strong> fibroblasts mayexhibit cytologic atypia, <strong>and</strong> some <strong>of</strong> <strong>the</strong>se cells maybe multinucleated, but mitosis is rare. Mast cells maybe numerous. There may be focal thrombosis, haemorrhage<strong>and</strong> chronic inflammatory reaction. With <strong>the</strong>advent <strong>of</strong> preoperative selective embolisation, iatrogenicemboli (Fig. 2.8b) are increasingly encounteredin resected specimens [232]. For more details on thistumour see Chap. 6.2.11 Malignant Sinonasal TumoursMalignant sinonasal tumours represent less than 1% <strong>of</strong>all cancers seen in humans <strong>and</strong> about 3% <strong>of</strong> all malignancies<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck region [160]. Despite <strong>the</strong>low rate <strong>of</strong> malignancy arising in <strong>the</strong> sinonasal tract, agreat variety <strong>of</strong> histological types <strong>of</strong> tumours may befound [216, 226]. The use <strong>of</strong> electron microscopy <strong>and</strong>more recent advances in immunohistochemistry <strong>and</strong>molecular biology have made it possible to refine <strong>the</strong>criteria for <strong>the</strong>ir correct recognition.Geographical differences in <strong>the</strong> relative frequency<strong>of</strong> certain histological types <strong>of</strong> malignant sinonasaltumours may be related to variations in <strong>the</strong> exposure


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 51Table 2.1. Malignant sinonasal tumours at <strong>the</strong> Hospital Clinic, University <strong>of</strong> Barcelona Medical SchoolHistologicaltype <strong>of</strong> tumourFrequencyn %Menn %Womenn %MeanageAgerangeSquamous cell carcinomaUndifferentiated carcinomaCylindrical cell carcinomaMalignant lymphomaMalignant melanomaHigh-grade adenocarcinomaAdenoid cystic carcinomaLow-grade adenocarcinomaOlfactory neuroblastomaMucoepidermoid carcinomaMalignant fibrous histiocytomaPlasmacytomaRhabdomyosarcomaMalignant schwannomaAdenosquamous carcinomaMyoepi<strong>the</strong>lial carcinomaKaposi’s sarcomaTeratocarcinosarcomaEwing’s sarcoma (PNET)54261919141311107444432221127139.59.57755322221.51110.50.538191515710743333212221–7073797950776440437575755033100100100100–167447346411122––––13027212150233660572525255067––––1006460595969595864365556513057664737762339–8741–8726–849–8956–8916–8122–6928–922–6750–6135–6550–658–5127–7061–7129–6634–40––Total 200 100 137 69 63 31 58 2–92to environmental carcinogens (see epidemiological aspectsSect. 11.1.1). In Table 2.1 <strong>the</strong> histological types <strong>of</strong>malignant sinonasal tumours diagnosed at <strong>the</strong> HospitalClinic <strong>of</strong> <strong>the</strong> University <strong>of</strong> Barcelona are presentedin decreasing order <strong>of</strong> frequency. The most frequenthistological types are: keratinising squamous cell carcinoma,undifferentiated carcinoma, cylindrical cellcarcinoma, malignant lymphoma, malignant melanoma,intestinal-type adenocarcinoma, adenoid cysticcarcinoma, low-grade adenocarcinomas <strong>and</strong> olfactoryneuroblastoma.A practical way to start classifying malignant sinonasaltumours is to separate <strong>the</strong>m initially into large <strong>and</strong>small cell categories. Among <strong>the</strong> large cell malignant tumours<strong>the</strong> most common types are: squamous cell carcinoma,cylindrical cell carcinoma, malignant melanoma,intestinal-type adenocarcinoma, <strong>and</strong> low-grade adenocarcinomas.To <strong>the</strong> most common small cell tumoursbelong <strong>the</strong> sinonasal undifferentiated carcinoma, malignantlymphoma, adenoid cystic carcinoma <strong>and</strong> olfactoryneuroblastoma. Large cell tumours account for approximately75% <strong>of</strong> <strong>the</strong> malignant sinonasal tumours <strong>and</strong> <strong>the</strong>small cell tumours for <strong>the</strong> remaining 25% [41].For staging <strong>of</strong> malignant sinonasal tumours <strong>the</strong> TNMclassification <strong>of</strong> 2002 <strong>and</strong> <strong>the</strong> TNM atlas <strong>of</strong> 2004 are recommended,since nasal cavity tumours are now included[237, 271a].2.11.1 Keratinising SquamousCell CarcinomaICD-O:8071/3At <strong>the</strong> nasal vestibule, keratinising squamous cell carcinomais <strong>the</strong> most common malignancy [130, 167, 245].Due to early recognition <strong>and</strong> easy access to treatment,<strong>the</strong>y usually have a more favourable prognosis than <strong>the</strong>ircounterpart <strong>of</strong> <strong>the</strong> sinonasal region.Sinonasal keratinising squamous cell carcinomarepresents up to 45–50% <strong>of</strong> <strong>the</strong> malignant tumours <strong>of</strong>this region in several series [93, 259]. At <strong>the</strong> HospitalClinic <strong>of</strong> <strong>the</strong> University <strong>of</strong> Barcelona, where non-keratinisingsquamous cell carcinomas (see Sect. 2.11.2on cylindrical cell carcinomas) are grouped separatelyfrom keratinising squamous cell carcinomas, <strong>the</strong> latteraccount for only 27% <strong>of</strong> <strong>the</strong> sinonasal malignancies.They predominate in males <strong>and</strong> <strong>the</strong> great majority areseen in patients aged over 50 years. The maxillary antrum,<strong>the</strong> lateral nasal wall <strong>and</strong> <strong>the</strong> sphenoidal sinusesare <strong>the</strong> most common sites, whereas <strong>the</strong> frontal <strong>and</strong>sphenoid sinuses are rarely involved. These tumoursgrow by local extension, infiltrating <strong>the</strong> neighbouringstructures, but lymph node metastases are rare [215].For neoplasms circumscribed to <strong>the</strong> nasal cavity <strong>the</strong>5-year survival is slightly above 50% [30], whereas inneoplasms <strong>of</strong> <strong>the</strong> maxillary antrum <strong>the</strong> 5-year survivalmay be as low as 25% [146].


52 A. Cardesa · L. Alos · A. Franchi2The occupational epidemiology <strong>of</strong> sinonasal squamouscell carcinoma has been strongly related to exposureto nickel [141, 243, 252, 253] <strong>and</strong> to a lesser extentto chromium, isopropyl alcohol <strong>and</strong> radium [218]. As ino<strong>the</strong>r territories <strong>of</strong> <strong>the</strong> respiratory tract, a definite associationbetween sinonasal squamous cell carcinoma <strong>and</strong> cigarettesmoking has been documented [26, 146]. Chronicsinonasal inflammation is considered a predisposingfactor. A case <strong>of</strong> carcinoma <strong>of</strong> <strong>the</strong> maxillary antrum afterthorotrast exposure has been reported [97]. Nitrosamines<strong>and</strong> to a lesser extent formaldehyde are strong nasal carcinogensin laboratory rodents [44, 155].Keratinising squamous cell carcinomas originate in<strong>the</strong> respiratory sinonasal mucosa from areas <strong>of</strong> pre-existingsquamous metaplasia <strong>and</strong> manifest <strong>the</strong> same range<strong>of</strong> histological appearances as those arising in o<strong>the</strong>r sites.They are characterised by <strong>the</strong> proliferation <strong>of</strong> malignantepi<strong>the</strong>lial cells with squamous differentiation <strong>and</strong> intercellularbridges. Malignancy is graded according to<strong>the</strong> degree <strong>of</strong> differentiation, cellular pleomorphism <strong>and</strong>mitotic activity. They are divided into well-differentiated,moderately differentiated <strong>and</strong> poorly differentiatedforms. Well-differentiated carcinomas are uncommonin this territory <strong>and</strong> when encountered need to be differentiatedfrom pseudoepi<strong>the</strong>liomatous types <strong>of</strong> hyperplasia<strong>and</strong> from verrucous carcinoma. Most conventionalkeratinising squamous cell carcinomas <strong>of</strong> <strong>the</strong> sinonasaltract present as moderately or poorly differentiatedtumours. Special types, such as verrucous carcinoma[104], spindle cell carcinoma [205, 276], basaloid-squamouscell carcinoma [16, 269] <strong>and</strong> adenosquamous carcinoma[10, 94] are occasionally found in <strong>the</strong> sinonasaltract. Regional lymph node involvement is seen in about17% <strong>of</strong> sinonasal squamous cell carcinomas <strong>and</strong> distantmetastases in about 1.5% [215].2.11.2 Cylindrical Cell CarcinomaICD-O:8121/3Cylindrical cell carcinoma, also known as non-keratinisingsquamous cell carcinoma, transitional cell carcinomaor schneiderian carcinoma, is a tumour composed <strong>of</strong> malignantproliferating cells derived from sinonasal respiratory(schneiderian) epi<strong>the</strong>lium. The name cylindrical cellcarcinoma was first coined by Ringertz in 1938 [212] <strong>and</strong>recommended as <strong>the</strong> preferred term by Shanmugaratnamin <strong>the</strong> WHO classification <strong>of</strong> 1991 [226].Grossly, <strong>the</strong> tumours grow in most cases as exophyticmasses showing ei<strong>the</strong>r a corrugated or a smooth surface.They may arise from <strong>the</strong> antrum, <strong>the</strong> lateral nasalwall or <strong>the</strong> ethmoid, <strong>the</strong> maxillary antrum being<strong>the</strong> most frequent site [193, 194]. They may occur concomitantlywith o<strong>the</strong>r non-neoplastic polypoid formations.Microscopically, cylindrical cell carcinoma iscomposed <strong>of</strong> papillary fronds (Fig. 2.9a), thick ribbons<strong>and</strong> polystratified masses <strong>of</strong> cells that give rise quite<strong>of</strong>ten to invaginations <strong>of</strong> <strong>the</strong> surface epi<strong>the</strong>lium, whichat low magnification may mimic inverted papilloma.The tumour cells are commonly cylindrical <strong>and</strong> havea tendency to form palisade arrangements perpendicularto <strong>the</strong> underlying basement membrane (Fig. 2.9b).The nuclei are atypical <strong>and</strong> show increased mitotic activity,as well as abnormal mitotic figures. The pattern<strong>of</strong> invasion is usually expansive, being characterised bypushing margins with focal infiltration <strong>of</strong> <strong>the</strong> stroma.The basement membrane remains in most cases conspicuous,despite stromal infiltration, which shouldnot be regarded as carcinoma in situ. Foci <strong>of</strong> squamousmetaplasia, with transition from cylindrical to squamousepi<strong>the</strong>lium, are not uncommon <strong>and</strong> when extensive<strong>the</strong>se tumours may be indistinguishable fromsquamous cell carcinoma. This resulted in denominationssuch as “transitional cell carcinoma” <strong>and</strong> “nonkeratinisingsquamous cell carcinoma”, which may beconfusing. The first because <strong>the</strong> term transitional hasalso been applied to carcinomas <strong>of</strong> <strong>the</strong> lymphoepi<strong>the</strong>lialtype, <strong>and</strong> <strong>the</strong> second due to <strong>the</strong> fact that tumours called“non-keratinising squamous cell carcinoma” also havefoci <strong>of</strong> keratinisation [267a]. In addition, <strong>the</strong> term cylindricalcell carcinoma should be preferred to non-keratinisingsquamous cell carcinoma because “pure” cylindricalcell carcinomas, without any squamous cellcomponent, carry a better prognosis than conventionalsquamous cell carcinomas [84]. Very recent observationssuggest a strong etiologic relationship <strong>of</strong> cylindricalcell carcinoma to high-risk human papillomavirus[69a]. A high proportion <strong>of</strong> <strong>the</strong>se tumours showmarked immunoreactivity for p16.More aggressive types <strong>of</strong> carcinoma, such as sinonasalundifferentiated carcinoma (SNUC) or high-gradeadenocarcinoma, may appear occasionally intermingledwith cylindrical cell carcinoma [226]. Two cases <strong>of</strong> cylindricalcell carcinoma with endodermal sinus-like featureshave been reported [162]. A full examination <strong>of</strong> <strong>the</strong>resected specimen is <strong>the</strong>refore m<strong>and</strong>atory before labellinga tumour a “pure cylindrical cell carcinoma”.The two main differential diagnoses <strong>of</strong> cylindricalcell carcinoma are <strong>the</strong> schneiderian papillomas <strong>of</strong> <strong>the</strong>inverted <strong>and</strong> oncocytic types, especially when <strong>the</strong>y haveconcomitant carcinomatous changes. Both types <strong>of</strong> papillomalack <strong>the</strong> atypical cellularity constantly seen in cylindricalcell carcinoma. When schneiderian papillomascoexist with cylindrical cell carcinomas, or with o<strong>the</strong>rtypes <strong>of</strong> carcinoma, <strong>the</strong> two components usually appeardemarcated from one ano<strong>the</strong>r although in contiguity.When <strong>the</strong> invaginating crypts <strong>of</strong> an inverted papillomaare filled with <strong>the</strong> cords <strong>and</strong> ribbons <strong>of</strong> a keratinisingor non-keratinising squamous cell carcinoma, <strong>the</strong> lesionrepresents a conventional squamous cell carcinomaarising in an inverted papilloma, which implies a worseprognosis than that <strong>of</strong> cylindrical cell carcinoma.


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 53abFig. 2.9. Cylindrical cell carcinoma. a Papillary structures coveredby malignant cells with cylindrical cytoplasm <strong>and</strong> basal nucleiarranged in palisades. b Marked immunoreactivity <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lialneoplastic cells for p16, which is related to high-risk HPV2.11.3 Sinonasal UndifferentiatedCarcinomaICD-O:8020/3Sinonasal undifferentiated carcinoma (SNUC) is definedas a high-grade malignant epi<strong>the</strong>lial neoplasm <strong>of</strong> <strong>the</strong> nasalcavity <strong>and</strong> paranasal sinuses, composed <strong>of</strong> small tomedium-sized cells, lacking evidence <strong>of</strong> squamous orgl<strong>and</strong>ular differentiation <strong>and</strong> <strong>of</strong> rosette formation [48,86, 114]. Cigarette smoking [86] <strong>and</strong> nickel exposure[252] have been associated with SNUC. Epstein-Barr virus(EBV) <strong>and</strong> <strong>the</strong> deletion <strong>of</strong> <strong>the</strong> retinoblastoma genehave been ruled out as factors involved in <strong>the</strong> development<strong>of</strong> this tumour [127]. Ionising radiation is ano<strong>the</strong>raetiologic factor, as radio<strong>the</strong>rapy ei<strong>the</strong>r for retinoblastomaor for nasopharyngeal carcinoma has been associatedwith SNUC [127].Sinonasal undifferentiated carcinoma occurs inboth sexes over a wide age range, with a median in <strong>the</strong>6th decade <strong>of</strong> life. It commonly originates from <strong>the</strong>ethmoidal region as a large fungating mass. Grossly,<strong>the</strong> tumours are quite frequently extensive lesions,obstructing <strong>the</strong> nasal cavity ei<strong>the</strong>r unilaterallyor bilaterally <strong>and</strong> invading <strong>the</strong> adjacent sinonasalstructures (Fig. 2.10a), as well as <strong>the</strong> orbit, skullbase <strong>and</strong> <strong>the</strong> brain. Microscopically, SNUC is composed<strong>of</strong> small to medium-sized, undifferentiatedcells, which arise via dysplastic changes from <strong>the</strong> basalcells <strong>of</strong> <strong>the</strong> surface epi<strong>the</strong>lium. The cells are polygonalwith distinct borders, showing round to oval, hyperchromaticor vesicular nuclei, with ei<strong>the</strong>r inconspicuousor slightly prominent nucleoli, surroundedby a moderate amount <strong>of</strong> ei<strong>the</strong>r amphophilic or eosinophiliccytoplasm (Fig. 2.10b). Mitotic figures arecommon. The tumour forms nests, cords <strong>and</strong> sheets<strong>of</strong> cells that show frequent areas <strong>of</strong> central necrosis<strong>and</strong> tendency to vascular (Fig. 2.10c) <strong>and</strong> perineuralinvasion. Ultrastructural studies demonstrate poorlyformed desmosomes in quite a number <strong>of</strong> cells, while<strong>the</strong> presence <strong>of</strong> tiny bundles <strong>of</strong> ton<strong>of</strong>ilaments is veryrare. Neurosecretory granules are very rarely found.SNUC are immunoreactive with epi<strong>the</strong>lial markers,such as CAM 5.2 <strong>and</strong> epi<strong>the</strong>lial membrane antigen(EMA). Variable reactivity can be seen with neuronspecificenolase (NSE), whereas <strong>the</strong>re is negative reactivityfor synaptophysin, chromogranin <strong>and</strong> o<strong>the</strong>rneuroendocrine markers. SNUC are also negative forEBV [48, 127].The two main differential diagnoses <strong>of</strong> SNUC aresmall cell (neuroendocrine) carcinoma (SCC) <strong>and</strong>high-grade olfactory neuroblastoma (ONB). All threeentities may share some clinical <strong>and</strong> light microscopicfeatures. However, SNUC <strong>and</strong> SCC show a markedimmunoreactivity for cytokeratins that is not seen inONB, <strong>and</strong> SNUC lacks <strong>the</strong> neuroendocrine immunoreactivityseen in SCC <strong>and</strong> ONB. Most lesions categorisedin <strong>the</strong> past as grade IV ONB are now consideredto be ei<strong>the</strong>r SNUC or SCC. This is important becauseSNUC <strong>and</strong> SCC have a worse prognosis than ONB. Inaddition, SNUC needs to be distinguished from o<strong>the</strong>rprimary sinonasal tumours, such as solid adenoid cysticcarcinoma, microcytic malignant melanoma, cylindricalcell carcinoma, primary sinonasal nasopharyngeal-typeundifferentiated carcinoma, lymphoma <strong>and</strong>o<strong>the</strong>rs (Table 2.2).Sinonasal undifferentiated carcinomas are very aggressivetumours. In most instances, <strong>the</strong> tumour is solarge <strong>and</strong> <strong>the</strong> infiltration is so extensive that completesurgical resection cannot be achieved. Radio<strong>the</strong>rapy<strong>and</strong> chemo<strong>the</strong>rapy are additional options, ei<strong>the</strong>r singleor combined. High-dose chemo<strong>the</strong>rapy <strong>and</strong> autologousbone marrow transplantation have been consideredas a form <strong>of</strong> treatment [241]. Prognosis <strong>of</strong> SNUCis dismal, with a median survival <strong>of</strong> 4 months to 1 year[86, 114]. In our experience survival after 2 years is lessthan 40%.


54 A. Cardesa · L. Alos · A. Franchi2abrequired for diagnosis [176]. Before placing a tumourwithin this category, a differential diagnosis <strong>of</strong> a primarytumour from <strong>the</strong> lung must be ruled out.This type <strong>of</strong> tumour has been well documented invarious head <strong>and</strong> neck territories, mainly in <strong>the</strong> parotidgl<strong>and</strong> <strong>and</strong> in <strong>the</strong> larynx. In <strong>the</strong> sinonasal tract, where<strong>the</strong>y are distinctly uncommon, small cell neuroendocrinecarcinomas have been less precisely characterised,<strong>and</strong> so far no unanimous consensus has been reachedwith regard to <strong>the</strong> way <strong>the</strong>y have to be separated fromo<strong>the</strong>r small cell tumours, ei<strong>the</strong>r round or undifferentiated,occurring in this region [51, 132, 151, 191, 208, 210,229, 264]. Table 2.2 provides <strong>the</strong> current criteria mostwidely accepted for <strong>the</strong>ir recognition.Small cell neuroendocrine carcinoma <strong>of</strong> <strong>the</strong> sinonasalregion is considered to derive from cells with neuroendocrinedifferentiation occasionally found in <strong>the</strong> seromucousgl<strong>and</strong>s. In some cases <strong>the</strong> tumour grows surrounding<strong>the</strong> seromucous gl<strong>and</strong>s <strong>of</strong> <strong>the</strong> lamina propria,as if it were originating from <strong>the</strong>m. They give rise tonests, cords <strong>and</strong> sheets <strong>of</strong> small, undifferentiated cells,with moulded nuclei <strong>and</strong> scanty cytoplasm. Immunohistochemistryexhibits a positive reaction for low molecularweight cytokeratins <strong>and</strong> EMA, as well as variablepositivity for neuron-specific enolase, Leu-7, CD56, synaptophysin<strong>and</strong> chromogranin. At least two neuroendocrinemarkers should demonstrate positivity [199]. Diligentsearching <strong>and</strong> expert h<strong>and</strong>s usually demonstrateneurosecretory granules by electron microscopy.Although its prognosis seems to be somewhat betterthan that <strong>of</strong> SNUC, or for similar tumours <strong>of</strong> <strong>the</strong> lung,small cell neuroendocrine carcinoma is a high-grademalignancy. Treatment should be a combination <strong>of</strong> surgery<strong>and</strong> radio<strong>the</strong>rapy, plus chemo<strong>the</strong>rapy.cFig. 2.10. Sinonasal undifferentiated carcinoma (SNUC) a Extensiveneoplastic growth invading ethmoid, orbit <strong>and</strong> sphenoid at<strong>the</strong> left <strong>and</strong> also <strong>the</strong> right ethmoid. Courtesy <strong>of</strong> Pr<strong>of</strong>. J. Traserra,Barcelona, Spain. b Nests <strong>of</strong> small to intermediate epi<strong>the</strong>lial cellsshowing markedly atypical nuclei <strong>and</strong> areas <strong>of</strong> necrosis. c Frequentfoci <strong>of</strong> intra-vascular invasion2.11.4 Small Cell (Neuroendocrine)CarcinomaICD-O:8041/3This is a high-grade malignant epi<strong>the</strong>lial tumour withhistological features similar to small cell carcinoma <strong>of</strong><strong>the</strong> lung [226]. Variable degrees <strong>of</strong> neuroendocrine differentiationmay be demonstrable by electron microscopyor immunohistochemistry, but have not always been2.11.5 Primary SinonasalNasopharyngeal-TypeUndifferentiated CarcinomaICD-O:8082/3Although nasopharyngeal carcinoma (NPC), alsoknown as lymphoepi<strong>the</strong>lioma, almost invariably arisesin <strong>the</strong> nasopharynx, “bona fide” primary sinonasal nasopharyngeal-typeundifferentiated carcinomas (PSN-PC) have recently been reported [127]. Due to <strong>the</strong> undifferentiatedappearance <strong>of</strong> cells in NPC <strong>and</strong> PSNPC,<strong>the</strong>se tumours may be lumped toge<strong>the</strong>r with SNUC ifunaware <strong>of</strong> <strong>the</strong>ir differences [48, 80, 127]. SNUC doesnot arise in <strong>the</strong> nasopharynx, but bulky lesions mayextend into this region. Also, NPC may extend from<strong>the</strong> nasopharynx into <strong>the</strong> sinonasal region. The distinctionbetween <strong>the</strong>se tumours can generally be madeon purely histological grounds, since SNUC lacks <strong>the</strong>lymphoplasmacytic cell infiltrate seen in most cases<strong>of</strong> NPC <strong>and</strong> PSNPC. Immunohistochemistry <strong>and</strong> in


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 55Table 2.2. Sinonasal undifferentiated tumors. Immunohistochemistry <strong>and</strong> genetics.CK NSE S-100 CG SYN NF EBV L MIC-2 t11;22AmplN-mycSNUC + ± – – – – – – – – –SCC + + – ± + – – – – – –PSNPC + – – – – – + – – – –SNML – – – – – – + + – – –PNET – + + ± + – – – + + –ONB – + (+) + + + – – – – –MNB – + – + + + – – – – +SNUC sinonasal undifferentiated carcinoma, SCC small cell (neuroendocrine) carcinoma, PSNPC primary sinonasal nasopharyngealtypecarcinoma, SNML sinonasal malignant lymphoma, PNET primitive neuroectodermal tumour, ONB olfactory neuroblastoma,MNB metastatic neuroblastoma, CK cytokeratin, NSE neuronal specific enolase, S-100 Protein S-100, CG chromogranin, SYN synaptophysin,NF neur<strong>of</strong>ilaments, EBV Epstein-Barr virus, L lymphoma markers, MIC-2 CD99, t(11;22) EWS-FLI1, Ampl amplification, (+)positive only in sustentacular cellssitu hybridisation are <strong>of</strong> great help in difficult cases.All three, NPC, PSNPC, <strong>and</strong> SNUC, react positively forlow molecular weight cytokeratins <strong>and</strong> EMA. In contrast,NPC <strong>and</strong> PSNPC are positive for EBV, whereasSNUC is negative. Until very recently, confusion <strong>of</strong>NPC <strong>and</strong> PSNPC with SNUC has led to <strong>the</strong> belief thatsome SNUC were related to EBV. The sharp distinction<strong>of</strong> <strong>the</strong>se entities is crucial because NPC <strong>and</strong> PSNPChave a better prognosis <strong>and</strong> are more responsive to radiation<strong>the</strong>rapy than SNUC.2.11.6 Malignant MelanomaICD-O:8720/3Sinonasal melanomas represent between 0.5 <strong>and</strong> 1.5% <strong>of</strong>all melanomas [25, 82, 157] <strong>and</strong> between 3 <strong>and</strong> 20% <strong>of</strong>sinonasal malignant neoplasms [25, 74]. They most frequentlydevelop after <strong>the</strong> fifth decade <strong>of</strong> life [25, 42, 250]<strong>and</strong> seem to originate from melanocytes present in <strong>the</strong>mucosa <strong>of</strong> <strong>the</strong> respiratory tract [25, 58, 275]. In our experience,it is not uncommon to see melanomas arising in anarea <strong>of</strong> squamous metaplasia . In contrast to Caucasians,black Africans <strong>of</strong>ten show visible pigmentation at sitescorresponding with <strong>the</strong> common locations <strong>of</strong> intranasalmelanomas, <strong>of</strong> which <strong>the</strong>y have a higher incidence [148].Although <strong>the</strong>re is not a significant sex predilection, menseem to be affected more than women [25, 29, 42]. Thesigns <strong>and</strong> symptoms <strong>of</strong> presentation <strong>of</strong> sinonasal melanomasare not specific. Epistaxis <strong>and</strong> nasal obstruction arefrequent when located in <strong>the</strong> nasal cavity.Grossly sinonasal malignant melanomas are ei<strong>the</strong>rpigmented (black-brown) or non-pigmented (pink-tan)lesions. In <strong>the</strong> nasal cavity, <strong>the</strong>y commonly arise in <strong>the</strong>anterior portion (Fig. 2.11a) <strong>of</strong> <strong>the</strong> septum <strong>and</strong> presentas tan-brown polypoid formations, with occasional ulcerated<strong>and</strong> hemorrhagic areas. When arising withinsinuses, <strong>the</strong>y present as extensive <strong>and</strong> widely infiltrativetumours. The development <strong>of</strong> an intranasal malignantmelanoma in an inverted papilloma has been reported[99].The histological features <strong>of</strong> sinonasal melanomasmay be as polymorphic as in <strong>the</strong>ir cutaneous counterpart.Metastatic disease needs to be ruled out, before<strong>the</strong>y are labelled as primary tumours. Primary melanomasmay be recognised by <strong>the</strong> presence <strong>of</strong> junctionalactivity (Fig. 2.11c) or by <strong>the</strong> finding <strong>of</strong> an intraepi<strong>the</strong>lialcomponent in <strong>the</strong> adjacent mucosa; never<strong>the</strong>less,<strong>the</strong>se features are usually lost in advanced stages <strong>of</strong> <strong>the</strong>disease. Histologically, melanomas are composed <strong>of</strong> mediumto large cells that may be polyhedral, round, fusiform(Fig. 2.11b), pleomorphic, microcytic, or a mixture<strong>of</strong> <strong>the</strong>m. Usually, <strong>the</strong>y have finely granular cytoplasm<strong>and</strong> nuclei with one or more eosinophilic nucleoli. Mitoticactivity is prominent. A rare balloon cell variantwith clear cytoplasm may mimic various types <strong>of</strong> clearcell tumours (see Chap. 5). Osteocartilaginous differentiationhas also been observed [244]. The cells <strong>of</strong> sinonasalmelanoma grow in solid, loosely cohesive, storiform,pseudo-alveolar or organoid patterns [25]. Two-thirds<strong>of</strong> sinonasal melanomas contain some intracytoplasmicbrown pigment (Fig. 2.11d) [25], which has to be confirmedas melanin by Masson-Fontana or Grimelius silverstains. However, in <strong>the</strong> sinonasal tract non-pigmentedmelanomas are not uncommon; in our Barcelona seriesup to 40% <strong>of</strong> <strong>the</strong> sinonasal melanomas are amelanotic.When melanin is scarce or is not found, diagnosismay be difficult, <strong>and</strong> special techniques are m<strong>and</strong>atory.Immunohistochemically, <strong>the</strong> cells <strong>of</strong> amelanotic melanomasare negative for cytokeratin <strong>and</strong> positive for vimentin,S-100 protein <strong>and</strong> HMB-45 [65, 82, 209], as well asanti-tyrosinase <strong>and</strong> o<strong>the</strong>r newly reported markers [207].Electron microscopy reveals <strong>the</strong> presence <strong>of</strong> pre-melanosomes<strong>and</strong>/or melanosomes.


56 A. Cardesa · L. Alos · A. Franchi2abcdFig. 2.11. Sinonasal malignant melanoma. a Darkly pigmentedpolypoid lesion <strong>of</strong> <strong>the</strong> anterior nasal cavity in contiguity with apigmented lesion <strong>of</strong> nasal skin. Courtesy <strong>of</strong> Pr<strong>of</strong>. J. Traserra, Barcelona,Spain. b Spindle cell non-pigmented malignant melanocyteswith storiform pattern. c Nests <strong>of</strong> non-pigmented, invasivemalignant melanocytes arising from metaplastic squamous epi<strong>the</strong>liumshowing junctional activity. d Pigmented malignant melanocytesunderneath ciliated respiratory epi<strong>the</strong>liumThe differential diagnosis <strong>of</strong> amelanotic malignantmelanoma <strong>of</strong> <strong>the</strong> sinonasal tract comprises a long list <strong>of</strong>entities. Epi<strong>the</strong>lioid melanomas mainly have to be distinguishedfrom non-keratinising squamous cell carcinomas,but also from o<strong>the</strong>r large cell carcinomas aswell as from epi<strong>the</strong>lioid malignant schwannomas [76]<strong>and</strong> from metastases. Microcytic melanoma may mimicSNUC <strong>and</strong> o<strong>the</strong>r small round cell tumours (Table 2.2).Spindle-cell melanoma may be mistaken for a variety <strong>of</strong>spindle-cell sarcomas (see Sects. 2.11.13 to 2.11.17).The prognosis <strong>and</strong> management is related to <strong>the</strong> peculiarbiology <strong>of</strong> <strong>the</strong> tumour. The prognostic significance<strong>of</strong> <strong>the</strong> level <strong>of</strong> local invasion, as established for cutaneousmelanomas, does not apply to mucosal melanomas because<strong>of</strong> <strong>the</strong> absence <strong>of</strong> histological l<strong>and</strong>marks analogousto <strong>the</strong> papillary <strong>and</strong> reticular dermis; never<strong>the</strong>less, inva-


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 57sion deeper than 0.5 mm is associated with decreased survival[25].Although many <strong>of</strong> <strong>the</strong> patients do not show initiallymph node involvement or disseminated metastases [25,83, 107] <strong>and</strong> have stage I disease at <strong>the</strong> time <strong>of</strong> initial diagnosis,<strong>the</strong> prognosis is bad due to a high recurrence rate[250]. This recurrence appears to be related to <strong>the</strong> multicentricity<strong>of</strong> <strong>the</strong> tumours <strong>and</strong> to <strong>the</strong> anatomic characteristics<strong>of</strong> <strong>the</strong> region that preclude adequate resection, whichis <strong>the</strong> treatment <strong>of</strong> choice [29, 256]. The utility <strong>of</strong> radio<strong>the</strong>rapyis controversial, but it can be <strong>of</strong> use in unresectablecases or to control recurrences [29, 95]. Immuno<strong>the</strong>rapy<strong>and</strong> chemo<strong>the</strong>rapy are also used for metastatic disease[256]. Five-year survival <strong>of</strong> sinonasal melanoma is reportedlyunder 35% [29, 250, 256]. The mean survival has notimproved during <strong>the</strong> past 15 years [32]. In our Barcelonaseries, <strong>the</strong> 5-year survival rate <strong>of</strong> 35% is similar to that<strong>of</strong> sinonasal squamous cell carcinoma. Patients with primarynasal melanomas had a significantly better 5-yearsurvival rate than patients with melanomas from o<strong>the</strong>rhead <strong>and</strong> neck sites [154].2.11.7 Olfactory NeuroblastomaICD-O:9522/3Olfactory neuroblastoma (ONB) is defined as a malignanttumour composed <strong>of</strong> neuroblasts derived from <strong>the</strong>olfactory membrane [14, 175, 246, 257]. Synonyms includeearly terminology such as es<strong>the</strong>sioneuroepi<strong>the</strong>lioma,es<strong>the</strong>sioneurocytoma <strong>and</strong> es<strong>the</strong>sioneuroblastoma.The site <strong>of</strong> origin <strong>of</strong> ONB is confined to <strong>the</strong> olfactorymucosa that lines <strong>the</strong> upper part <strong>of</strong> <strong>the</strong> nasal cavity [181].On rare occasions ONB predominantly involves <strong>the</strong> superioraspect <strong>of</strong> <strong>the</strong> cribiform plate <strong>and</strong> grow as intracranialmasses [15, 186]. Before establishing a diagnosis<strong>of</strong> <strong>the</strong> extremely rare entity known as “ectopic” olfactoryneuroblastoma, which implies absence <strong>of</strong> involvement<strong>of</strong> <strong>the</strong> olfactory membrane, o<strong>the</strong>r sinonasal small roundcell tumours have to be carefully ruled out (Table 2.2).Olfactory neuroblastoma has a bimodal age distributionwith peaks in <strong>the</strong> 2nd <strong>and</strong> 6th decades [70]. Thisclearly differs from adrenal neuroblastoma, with mostcases arising in children under 4 years <strong>of</strong> age. Both sexesare equally affected. Nasal obstruction, rhinorrhoea <strong>and</strong>epistaxis are <strong>the</strong> most common presenting symptoms.Grossly, <strong>the</strong> tumours are <strong>of</strong>ten unilateral, presentingas smooth polypoid or fungating masses <strong>of</strong> fleshy consistency<strong>and</strong> yellow to pink colour (Fig. 2.12a).At low magnification, ONB exhibits one <strong>of</strong> two mainpatterns <strong>of</strong> growth [176]. Most <strong>of</strong>ten, it presents a lobulararrangement with well-defined groups <strong>of</strong> tumourcells separated by abundant oedematous stroma. Less frequently,<strong>the</strong> tumour grows as diffuse sheets <strong>of</strong> cells withscanty, but highly vascular stroma. The neoplastic neuroblastsare typically small, showing round to oval nucleiwith stippled chromatin, absent or small nucleoli, <strong>and</strong>minimal cytoplasm. They are commonly separated by aneur<strong>of</strong>ibrillary matrix formed by neuronal cell processesin which axons may be demonstrable by conventionalsilver stains. This background, seen in about 85% <strong>of</strong>ONBs, is <strong>the</strong> most helpful diagnostic feature (Fig. 2.12b).The Homer-Wright type <strong>of</strong> rosettes is quite characteristic<strong>of</strong> ONB; however, <strong>the</strong>y are less commonly seen. Theyform when <strong>the</strong> tumour cells surround <strong>the</strong> neur<strong>of</strong>ibrillarymatrix in collar-like arrangements. Even more rareare <strong>the</strong> true olfactory Flexner-Wintersteiner type <strong>of</strong> rosettes,which depict well-defined lumina lined by columnarcells resembling olfactory epi<strong>the</strong>lium. These cells generallyhave basally located nuclei <strong>and</strong> merge with <strong>the</strong> adjacentneuroblasts without any intervening basal lamina.Perivascular pseudorosettes, formed by tumour cells arrangedaround capillaries, are <strong>of</strong> no diagnostic value, for<strong>the</strong>y may be found in several types <strong>of</strong> neoplasms.The scheme proposed by Hyams [122] to grade ONBinto four groups carries diagnostic <strong>and</strong> prognostic implications(Table 2.3). The differential diagnosis <strong>of</strong> ONBincludes a wide variety <strong>of</strong> small round cell tumours arisingin <strong>the</strong> sinonasal region (Table 2.2). Immunohistochemically,ONB shows diffuse positivity for NSE <strong>and</strong>synaptophysin, but is less <strong>of</strong>ten positive for chromogranin.In tumours with a nesting pattern, sustentacularcells are positive for S-100 protein, but generally negativefor cytokeratin, although in ONB with a nestingpattern a few tumours may exhibit focal staining for lowmolecular weight cytokeratins. They are negative forEMA. Neur<strong>of</strong>ilament protein <strong>and</strong> o<strong>the</strong>r markers <strong>of</strong> neuraldifferentiation are more <strong>of</strong>ten expressed in tumourswith diffuse, sheet-like patterns [53, 87, 246, 268]. Electronmicroscopy shows evidence <strong>of</strong> neuroblastic differentiation,demonstrating neuritic processes, neurotubules<strong>and</strong> membrane-bound dense-core granules [131,159, 247]. The human analogue <strong>of</strong> achaete-scute gene(HASH1), expressed in immature olfactory neurons,is also expressed in ONB [45]. Conversely, <strong>the</strong> olfactorymarker protein [182], expressed exclusively in matureolfactory neurons, is not. ONB lacks CD 99 (MIC-2) expression, as well as <strong>the</strong> t(11;22) translocation characteristic<strong>of</strong> primary neuroectodermal tumour (PNET)[13, 263]. It also lacks <strong>the</strong> molecular genetic changes <strong>of</strong>sympa<strong>the</strong>tic neuroblastoma, which, in children, may bemetastatic to <strong>the</strong> sinonasal region.Staging <strong>of</strong> ONB is based on <strong>the</strong> Kadish system [129], inwhich stage A disease is confined to <strong>the</strong> nasal cavity, stageB to <strong>the</strong> nasal cavity <strong>and</strong> paranasal sinuses, <strong>and</strong> stage Cshows local or distant spread beyond <strong>the</strong> nasal cavity orsinuses. This correlates with prognosis [70]. Necrosis is<strong>the</strong> single histological feature that seems to correlate withpoor survival [175]. About two-thirds <strong>of</strong> recurrences arein <strong>the</strong> form <strong>of</strong> local disease, whereas locoregional recurrences,with intracranial extension or involvement <strong>of</strong>cervical lymph nodes, represent about 20%, <strong>and</strong> distant


58 A. Cardesa · L. Alos · A. Franchi2<strong>of</strong> uniform, small, undifferentiated, primitive neuroectodermalcells (Fig. 2.13a) [149]. The great majority<strong>of</strong> <strong>the</strong>se tumours will react strongly with antibodiesagainst <strong>the</strong> MIC-2 (CD99) protein product (Fig. 2.13b).This marker is <strong>of</strong> considerable value, but it is by nomeans specific. A growing number <strong>of</strong> o<strong>the</strong>r neoplasmsexpressing this protein have been documented. Among<strong>the</strong>se are T-cell lymphomas [263]. The st<strong>and</strong>ard translocationt(11; 22) (q24; q12) <strong>of</strong> PNET [260] results in<strong>the</strong> fusion <strong>of</strong> <strong>the</strong> EWS-FLI1 genes. The detection <strong>of</strong> <strong>the</strong>chimeric transcript by techniques <strong>of</strong> molecular biologyconfirms <strong>the</strong> diagnosis [238]. We have seen one example<strong>of</strong> PNET arising from <strong>the</strong> maxillary antrum [39],which ultrastructurally showed rudimentary neuriticdifferentiation, as well as scanty microtubule formation.This raised <strong>the</strong> differential diagnostic dilemma<strong>of</strong> “ectopic olfactory neuroblastoma”; never<strong>the</strong>less, <strong>the</strong>tumour cells were positive for MIC-2 <strong>and</strong> showed <strong>the</strong>t(11;22)(q24;q12) translocation, findings that are characteristicallynegative in ONB [13].a2.11.9 High-GradeSinonasal Adenocarcinomas2.11.9.1 Intestinal-TypeAdenocarcinomabFig. 2.12. a Olfactory neuroblastoma: polypoid mass <strong>of</strong> fleshyconsistency <strong>and</strong> pink colour. b Well-differentiated olfactory neuroblastoma:nests <strong>of</strong> small round neuroblasts embedded in a neur<strong>of</strong>ibrillarymatrix are surrounded by delicate networks <strong>of</strong> capillaryblood vesselsmetastases account for <strong>the</strong> rest [70]. Distant metastasesmainly involve bone <strong>and</strong> lung [68]. Complete surgical excision,<strong>of</strong>ten followed by radiation <strong>the</strong>rapy <strong>and</strong>/or chemo<strong>the</strong>rapy,seems to be <strong>the</strong> treatment <strong>of</strong> choice [14, 179]. Inadvanced ONB, high-dose chemo<strong>the</strong>rapy <strong>and</strong> autologousbone marrow transplantation has been used [68, 188].2.11.8 PrimitiveNeuroectodermal TumourICD-O:9364/3Approximately 9% <strong>of</strong> extraosseous Ewing’s sarcoma/primitive neuroectodermal tumour (PNET) arise in <strong>the</strong>head <strong>and</strong> neck region [176]. This tumour is composedICD-O:8144/3This is a tumour with histological features resemblingcolorectal adenocarcinoma [126, 220]. It is considered tooriginate through intestinal metaplasia <strong>of</strong> <strong>the</strong> ciliated respiratorycells lining <strong>the</strong> schneiderian membrane. Thistumour is <strong>the</strong> most common type <strong>of</strong> sinonasal adenocarcinoma,representing about 6–13% <strong>of</strong> malignanciesdeveloping in <strong>the</strong> sinonasal tract [41, 105, 216]. Metastasisfrom gastrointestinal adenocarcinoma should beruled out before a tumour is labelled as a primary <strong>of</strong> thisregion. These tumours are strongly associated with exposureto different types <strong>of</strong> dust, mainly hardwood, butalso s<strong>of</strong>twood dusts, as well as lea<strong>the</strong>r dust [4, 5, 47, 102,123, 124, 142]. About 20% <strong>of</strong> sinonasal intestinal-typeadenocarcinomas seem to be sporadic, without evidence<strong>of</strong> exposure to industrial dusts [4].Grossly, <strong>the</strong>y have a fungating appearance with ei<strong>the</strong>rpolypoid or papillary features. Occasionally, <strong>the</strong>y mayhave a gelatinous consistency resembling a mucocele. Themost common location is <strong>the</strong> ethmoidal region [17]. Histologically,<strong>the</strong> tumour is mainly composed <strong>of</strong> columnarmucin-secreting cells <strong>and</strong> goblet cells [17]. Some well-differentiatedtumours may also contain resorptive cells, argentaffincells <strong>and</strong> Paneth cells. Endocrine-amphicrineenteric differentiation may occasionally be found [222].Metaplastic <strong>and</strong> atypical changes have been observed inadjacent pre-neoplastic epi<strong>the</strong>lium [270]. These tumours


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 59Table 2.3. Olfactory neuroblastoma. Hyams Grading Scheme [122].Histologic grades 1 2 3 4Lobular architecture Present Present ± +Mitotic activity Absent Present Prominent MarkedNuclear pleomorphism Absent Moderate Prominent MarkedFibrillary matrix Prominent Moderate Slight AbsentRosettes H-W + H-W + Flexner + AbsentNecrosis Absent Absent Occasional CommonCalcification ± + Absent AbsentH-W Homer-Wright rosettes, + present or absentabFig. 2.13. Primitive neuroectodermal tumour (PNET). a Monotonousproliferation <strong>of</strong> small, round, undifferentiated cells. bCD99-positive immune reaction at <strong>the</strong> cellular membranedepict different histological patterns that may be predominantlypapillary, gl<strong>and</strong>ular, compact, mucinous or mixed[17, 23]. Papillary tumours mainly consist <strong>of</strong> elongatedoutgrowths lined by intestinal-type cells with markedlyatypical pseudostratified nuclei (Fig. 2.14a). Althoughmost <strong>of</strong> <strong>the</strong>m are high-grade tumours, low-grade forms(Fig. 2.14b) mimicking colonic villous adenoma may occasionallyoccur [174]. The gl<strong>and</strong>ular pattern resemblescommon-type intestinal adenocarcinoma. Compact orsolid forms show poorly differentiated nests <strong>of</strong> cells inwhich gl<strong>and</strong>ular formation is rarely seen. In <strong>the</strong> mucinouspattern, more than 50% <strong>of</strong> <strong>the</strong> tumour is composed<strong>of</strong> dilated mucin-filled gl<strong>and</strong>s lined by columnar mucin-secretingepi<strong>the</strong>lium, <strong>and</strong> lakes <strong>of</strong> mucin containingfragmented epi<strong>the</strong>lial elements (Fig. 2.14c). O<strong>the</strong>r mucinoustumours show mucin-filled cells with <strong>the</strong> pattern <strong>of</strong>“signet-ring” cell carcinoma. Various attempts have beenmade to correlate histopathological grading <strong>and</strong> typingwith clinical behaviour [78, 81, 140].Features such as cytologic atypia, high mitotic rate <strong>and</strong>areas <strong>of</strong> necrosis, which are common findings in most intestinal-typeadenocarcinomas, help to distinguish <strong>the</strong>high-grade variants from rare low-grade intestinal-typeadenocarcinomas <strong>and</strong> from mucoceles. The lack <strong>of</strong> epidermoid<strong>and</strong> squamous differentiation separates <strong>the</strong>se tumoursfrom mucoepidermoid <strong>and</strong> adenosquamous carcinomas.Immunohistochemistry <strong>and</strong> electron microscopyhave confirmed <strong>the</strong> enteric differentiation <strong>of</strong> <strong>the</strong> tumourcells [24]. These tumours are positive for a wide spectrum<strong>of</strong> cytokeratin markers, whereas <strong>the</strong>y are only occasionallypositive for carcinoembryonic antigen [166]. Intestinal-typeadenocarcinomas are frequently but not alwayspositive for cytokeratin 7, while most express cytokeratin20 <strong>and</strong> CDX-2, two markers related to intestinal differentiation[79]. The prognosis for high-grade intestinal-typeadenocarcinoma is poor. Recurrences <strong>and</strong> subsequentdeeply invasive local growth are frequent; however,lymph node <strong>and</strong> distant metastases are rare [17, 78,142]. Treatment <strong>of</strong> choice is complete surgical resectionfollowed by radio<strong>the</strong>rapy.2.11.9.2 Salivary-TypeHigh-Grade AdenocarcinomasAdenoid Cystic CarcinomaICD-O:8200/3Adenoid cystic carcinoma (see Chap. 5) is <strong>the</strong> most commonmalignant salivary-type <strong>of</strong> tumour <strong>of</strong> <strong>the</strong> upper


60 A. Cardesa · L. Alos · A. Franchi2aggressive tumour from <strong>the</strong> seromucous gl<strong>and</strong>s <strong>of</strong> <strong>the</strong>upper respiratory tract may occasionally occur. We haveseen one example <strong>of</strong> SDC originating in <strong>the</strong> maxillaryantrum, in which <strong>the</strong> characteristic ductal pattern, withcomedo-type necrosis, was only evident in <strong>the</strong> metastasesto <strong>the</strong> subm<strong>and</strong>ibular lymph nodes. The primarytumour was initially classified as adenocarcinoma noto<strong>the</strong>rwise specified (NOS) due to <strong>the</strong> absence <strong>of</strong> convincingdiagnostic features [40]. Carcinoma ex pleomorphicadenoma may also occur in <strong>the</strong> sinonasal tract [110].ab2.11.10 Low-Grade SinonasalAdenocarcinomasLow-grade adenocarcinomas arising primarily within<strong>the</strong> sinonasal tract are an uncommon <strong>and</strong> heterogeneousgroup <strong>of</strong> tumours [23, 113, 139, 150]. Some <strong>of</strong><strong>the</strong>se neoplasms show apparent histological continuitywith <strong>the</strong> normal surface epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong> sinonasalmucosa, whereas o<strong>the</strong>rs are <strong>of</strong> salivary gl<strong>and</strong> origin. Allhave better prognosis <strong>and</strong> different clinical presentationthan <strong>the</strong>ir high-grade counterpart. With <strong>the</strong> exception<strong>of</strong> <strong>the</strong> well-differentiated, low-grade, adenocarcinomas<strong>of</strong> intestinal type, no correlation with occupational activitieshas been found in <strong>the</strong>se tumours.2.11.10.1 Non-Salivary-TypeLow-Grade AdenocarcinomascFig. 2.14. Intestinal-type adenocarcinoma. a Papillary outgrowth<strong>of</strong> intestinal-like malignant epi<strong>the</strong>lium. Destruction <strong>of</strong> sinonasalbone at <strong>the</strong> bottom. b Low-grade variant mimicking villous adenoma<strong>of</strong> <strong>the</strong> colon. c Mucinous variant mimicking mucocelerespiratory tract <strong>and</strong> constitutes 5–10% <strong>of</strong> all sinonasalmalignancies [43, 105, 206]. It is most common in <strong>the</strong>maxillary antrum, followed by <strong>the</strong> nasal cavity [60], althoughethmoid, sphenoid <strong>and</strong> frontal sinuses may alsobe involved [110, 169, 255].O<strong>the</strong>r Salivary-Type High-Grade AdenocarcinomasWith <strong>the</strong> exception <strong>of</strong> adenoid cystic carcinoma, <strong>the</strong>setumours are quite rare in <strong>the</strong> sinonasal region. Althoughmost salivary duct carcinomas (SDCs) arise from <strong>the</strong>major salivary gl<strong>and</strong>s, <strong>the</strong> development <strong>of</strong> this highlyNon-salivary-type low-grade sinonasal adenocarcinomasare a distinctive group <strong>of</strong> tumours comprising well-differentiatedtubular or papillary structures, or a combination<strong>of</strong> both. They are lined by a single layer <strong>of</strong> cuboidalor columnar cells that display uniform round or ovalnuclei, inconspicuous nucleoli, minimal cytologic atypia<strong>and</strong> scarce mitotic figures. They are locally infiltrative<strong>and</strong> have a tendency towards local recurrence [113].Different histological patterns may be recognised:papillary, gl<strong>and</strong>ular, mucinous, trabecular, cribriform<strong>and</strong> clear cell. The papillary pattern is characterised bycomplex papillary fronds lined by bl<strong>and</strong> columnar cellsthat may occasionally mimic oncocytic (columnar) cellpapilloma. Quite similar tumours also develop in <strong>the</strong>nasopharynx [267]. The gl<strong>and</strong>ular pattern may simulateadenoma; never<strong>the</strong>less, <strong>the</strong> presence <strong>of</strong> closely packedgl<strong>and</strong>s, forming back-to-back arrangements, indicates<strong>the</strong> true malignant nature. Mucinous tumours have to bedistinguished from mucoceles <strong>and</strong> from mucinous adenocarcinoma<strong>of</strong> intestinal or salivary type [20, 224]. Thetrabecular pattern may resemble acinic cell carcinoma[200]. The cribriform arrangements have to be distinguishedfrom low-grade salivary duct carcinoma <strong>of</strong> salivarygl<strong>and</strong>s [62]. The clear cell type has to be separatedfrom <strong>the</strong> salivary-type tumours with clear cells <strong>and</strong>


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 61Fig. 2.15. Tubulopapillary carcinoma: low-grade proliferation <strong>of</strong>cuboidal to columnar epi<strong>the</strong>lial cells forming tubules at <strong>the</strong> centre<strong>and</strong> papillae at <strong>the</strong> surfaceafrom metastatic renal cell carcinoma [31, 280]. A tubulopapillaryvariant has recently been reported (Fig. 2.15)[234] that has to be differentiated from terminal tubulusadenocarcinoma <strong>of</strong> <strong>the</strong> nasal seromucous gl<strong>and</strong>s [139].2.11.10.2 Salivary-Type Low-GradeAdenocarcinomasMucoepidermoid carcinoma, polymorphous low-gradeadenocarcinoma (Fig. 2.16) <strong>and</strong> acinic cell carcinomaoriginate only on rare occasions from <strong>the</strong> seromucousgl<strong>and</strong>s <strong>of</strong> <strong>the</strong> sinonasal mucosa [43, 110, 150, 190, 200,239]. Most mucoepidermoid carcinomas <strong>of</strong> <strong>the</strong> sinonasaltract are low-grade. Some large oncocytic tumours<strong>of</strong> <strong>the</strong> sinonasal tract may behave in a locally aggressivefashion <strong>and</strong> are better classified as low-grade adenocarcinomas[55, 103, 110]. All <strong>the</strong>se tumours are dealt within detail in Chap. 5 on salivary gl<strong>and</strong>s. Their main differentialdiagnoses are o<strong>the</strong>r salivary- or non-salivarytypelow-grade adenocarcinomas.2.11.11 Sinonasal Malignant LymphomasMalignant lymphomas <strong>of</strong> <strong>the</strong> sinonasal region compriseapproximately 6% <strong>of</strong> all sinonasal malignancies [134]. Inour Barcelona series, <strong>the</strong>y account for 9.5% (Table 2.1).In western countries, about 50% <strong>of</strong> sinonasal lymphomasare <strong>of</strong> B-cell type, whereas <strong>the</strong> o<strong>the</strong>r 50% mostly showedNK/T-cell lineage [38]; never<strong>the</strong>less, o<strong>the</strong>r reports pointto more variable rates [3, 72, 77, 85]. Conversely, in orientalpopulations most primary lymphomas <strong>of</strong> <strong>the</strong> nasalcavity <strong>and</strong> nasopharynx are <strong>of</strong> NK/T cell lineage [49, 50,52, 92, 233].Sinonasal B-cell lymphomas are in general composed<strong>of</strong> a diffuse proliferation <strong>of</strong> large lymphoid cells, or <strong>of</strong> abFig. 2.16. Polymorphous low-grade adenocarcinoma. a CT scanshowing an irregularly nodular lesion destroying <strong>the</strong> anterior nasalseptum. Courtesy <strong>of</strong> Pr<strong>of</strong>. J. Traserra, Barcelona, Spain. b Variegatedgl<strong>and</strong>ular arrangements composed <strong>of</strong> tubules with bl<strong>and</strong>cellularity are seen beneath respiratory epi<strong>the</strong>liumdiffuse mixed pattern <strong>of</strong> small <strong>and</strong> large cells. They infiltrate<strong>and</strong> exp<strong>and</strong> <strong>the</strong> subepi<strong>the</strong>lial s<strong>of</strong>t tissue <strong>and</strong> mayextend into <strong>the</strong> underlying bone. Sinonasal B-cell lymphomaslack epi<strong>the</strong>liotropism, polymorphous cell infiltrate,angiocentricity, prominent necrosis, <strong>and</strong> fibrosis.They are usually positive for B-cell markers (CD20<strong>and</strong> CD79a) <strong>and</strong> negative for NK/T cell markers. lightchain restriction is seen more <strong>of</strong>ten than restriction.They are <strong>of</strong>ten negative for EBV markers. Radio<strong>the</strong>rapy<strong>and</strong> chemo<strong>the</strong>rapy is <strong>the</strong> st<strong>and</strong>ard treatment for advancedtumours [213].Sinonasal NK/T cell lymphomas were labelled in pastdecades with terms such as “lethal midline granuloma”,“polymorphic reticulosis” <strong>and</strong> “angiocentric T-cell lymphoma”,among o<strong>the</strong>rs. Until quite recently, non-B cellsinonasal lymphomas were considered as o<strong>the</strong>r forms <strong>of</strong>T-cell lymphoma, frequently displaying angiocentricity.Patients may present ei<strong>the</strong>r with an obstructive massor with mid-facial destructive lesions. Histologically, anangiocentric <strong>and</strong> angiodestructive infiltrate with extensivenecrosis (Fig. 2.17a) is frequently seen. In NK/T-celllymphoma, cells may be small, medium-sized, large, oranaplastic, <strong>and</strong> may show a conspicuous admixture <strong>of</strong>


62 A. Cardesa · L. Alos · A. Franchi2abFig. 2.17. Sinonasal NK/T lymphoma. a Angiocentric infiltrate<strong>of</strong> atypical lymphocytes with extensive necrotic areas. b Stronglypositive CD56 immunoreactioninflammatory cells. Pseudoepi<strong>the</strong>liomatous hyperplasia<strong>of</strong> <strong>the</strong> covering epi<strong>the</strong>lium may occur [49]. NK/T celllymphoma is almost always associated with EBV positivity.The most typical immunophenotypes are CD2+,CD56+ (Fig. 2.17b), surface CD3–, <strong>and</strong> cytoplasmicCD3epsilon+. Most cases are also positive for cytotoxicgranule-associated proteins (granzyme B, TIA-1 <strong>and</strong>perforin). They are usually negative for o<strong>the</strong>r T <strong>and</strong> NKcellassociated markers. Sinonasal lymphomas demonstratingCD3epsilon+, CD56–, cytotoxic molecule+, <strong>and</strong>EBV+ are also included within <strong>the</strong> NK/T category. Thecommonest cytogenetic abnormality found in NK/Tcelllymphoma is <strong>the</strong> 6q-22-23 deletion [233]. The prognosis<strong>of</strong> nasal NK/T-cell lymphoma is variable. Some patientsrespond well to <strong>the</strong>rapy <strong>and</strong> o<strong>the</strong>rs die <strong>of</strong> diseasedespite aggressive <strong>the</strong>rapy [52].Sinonasal malignant lymphomas <strong>of</strong> ei<strong>the</strong>r B-cell or T-cell derivation need a careful differential diagnosis witho<strong>the</strong>r small round cell tumours (Table 2.2) <strong>and</strong> with extramedullaryplasmacytoma [6, 46], as well as with extramedullarytumours composed <strong>of</strong> myeloid or lymphoidblasts [133].2.11.12 Extramedullary PlasmacytomaICD-O:9731/3Plasmacytoma <strong>of</strong> <strong>the</strong> sinonasal tract appears as a diffuseinfiltration <strong>of</strong> mature plasma cells <strong>of</strong> <strong>the</strong> mucosa;occasionally, tumour cells are less differentiated, <strong>and</strong>diagnosis may be difficult on a histologic basis [2, 6, 46,185]. Immunohistochemical staining for CD138 <strong>and</strong> <strong>and</strong> chains may be helpful. Full examination <strong>of</strong> <strong>the</strong>patient is required to exclude disseminated disease.2.11.13 FibrosarcomaICD-O:8810/3Fibrosarcoma <strong>of</strong> <strong>the</strong> sinonasal tract occurs across awide age range, most commonly causing obstruction orepistaxis. The histologic appearance is that <strong>of</strong> a spindlecell lesion, with fascicles or bundles <strong>of</strong> neoplastic cellsintersecting at various angles, sometimes with a herringbonepattern. Most sinonasal fibrosarcomas have alow-grade appearance, with moderate cellularity <strong>and</strong> alow mitotic rate [111]. In accordance, <strong>the</strong> behaviour ismore <strong>of</strong>ten characterised by repeated local recurrences,while distant metastases are rare. The differentialdiagnosis includes desmoid fibromatosis, leiomyosarcoma,nerve sheath tumours, spindle cell carcinoma<strong>and</strong> melanoma.2.11.14 Malignant FibrousHistiocytomaICD-O:8830/3Malignant fibrous histiocytoma is a high-grade sarcoma<strong>of</strong> adulthood histologically consisting <strong>of</strong> a proliferation<strong>of</strong> spindle cells arranged in a storiform pattern, intermixedwith atypical pleomorphic, <strong>of</strong>ten multinucleatedgiant cells. In <strong>the</strong> sinonasal tract it presents as a highlyaggressive <strong>and</strong> destructive lesion, with bone invasion<strong>and</strong> extension in adjacent structures [201]. Before a diagnosis<strong>of</strong> malignant fibrous histiocytoma is rendered,o<strong>the</strong>r pleomorphic malignant tumours, like leiomyosarcoma,osteosarcoma <strong>and</strong> sarcomatoid carcinoma shouldbe excluded by means <strong>of</strong> immunohistochemical or ultrastructuralanalysis.


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 632.11.15 LeiomyosarcomaICD-O:8890/3Leiomyosarcoma <strong>of</strong> <strong>the</strong> sinonasal tract is an extremelyrare neoplasm, with identical histological appearance<strong>and</strong> immunopr<strong>of</strong>ile to <strong>the</strong> s<strong>of</strong>t tissue counterpart [91]. If<strong>the</strong> tumour is limited to <strong>the</strong> nasal cavity <strong>the</strong> prognosis isgood, <strong>and</strong> according to Kuruvilla et al. [144], sinonasalleiomyosarcoma can be regarded as a locally aggressiveneoplasm with limited metastatic potential that shouldbe treated by surgery alone. Distinction from sinonasalleiomyoma is based on mitotic activity <strong>and</strong> cellularatypia.2.11.16 RhabdomyosarcomaICD-O:8900/3Rhabdomyosarcoma is <strong>the</strong> most common sinonasal malignancy<strong>of</strong> <strong>the</strong> paediatric age, but it is also observed inadults [37, 116]. The most common histologic subtypesare <strong>the</strong> embryonal <strong>and</strong> <strong>the</strong> alveolar [37]. The overall 5-year survival is around 40%; adult age <strong>and</strong> alveolar subtypeare adverse prognostic factors. Treatment includesa combination <strong>of</strong> radio<strong>the</strong>rapy <strong>and</strong> chemo<strong>the</strong>rapy, withsurgical resection reserved for residual disease. The risk<strong>of</strong> neck involvement is high.2.11.17 Malignant PeripheralNerve Sheath TumourICD-O:9560/3Malignant peripheral nerve sheath tumour (MPNST) <strong>of</strong><strong>the</strong> sinonasal tract is a very rare neoplasm [163] that isprobably under-recognised due to <strong>the</strong> lack <strong>of</strong> reproduciblehistologic criteria <strong>and</strong> to <strong>the</strong> tendency <strong>of</strong> <strong>the</strong>se tumoursto be negative for <strong>the</strong> commonly used immunohistochemicalmarkers <strong>of</strong> nerve sheath differentiation.Only in some cases can <strong>the</strong> diagnosis be based on <strong>the</strong>identification <strong>of</strong> a pre-existing neur<strong>of</strong>ibroma. Histologically,MPNST is a moderately to highly cellular spindlecell proliferation, with variable mitotic activity <strong>and</strong> areas<strong>of</strong> necrosis. A variant composed <strong>of</strong> epi<strong>the</strong>lioid cells hasbeen described in <strong>the</strong> sinonasal cavities (Fig. 2.18) [76].Some tumours may show morphologic <strong>and</strong> immunohistochemicalfeatures <strong>of</strong> skeletal muscle differentiation<strong>and</strong> are designated “malignant triton tumours” [138].2.11.18 TeratocarcinosarcomaICD-O:8980/3The term “ teratocarcinosarcoma” is designated to anunusual entity in which a malignant sinonasal teratomatoustumour also shows features <strong>of</strong> carcinosarcomaFig. 2.18. Sinonasal malignant epi<strong>the</strong>lioid schwannoma: markedS-100 protein positivity in a large cell malignant neoplasm, mimickingamelanotic melanoma[112]. Patients suffering from this malignancy are exclusivelyadults (age range 18–79 years; mean 60 years) [64,75, 112, 156, 195, 227]. There is a male predominance<strong>and</strong> symptoms are non-specific with nasal obstruction<strong>and</strong> epistaxis produced by a nasal cavity mass (Fig. 2.19a)[112].Histologically, sinonasal teratocarcinosarcoma(SNTCS) comprises a multiplicity <strong>of</strong> cell types <strong>of</strong>varying maturity (Fig. 2.19b). The epi<strong>the</strong>lial componentincludes keratinising squamous epi<strong>the</strong>lium, pseudostratifiedcolumnar ciliated epi<strong>the</strong>lium <strong>and</strong> gl<strong>and</strong>ularstructures lined by cuboidal <strong>and</strong> columnar cellsincluding mucus cells. Masses <strong>of</strong> immature epi<strong>the</strong>lialcells, some containing glycogen or mucin, are frequentlyfound. “Foetal-appearing” clear cell squamousepi<strong>the</strong>lium is a common finding <strong>and</strong> a very importantdiagnostic clue for some authors [112]. Neuroepi<strong>the</strong>lialelements with rosettes <strong>and</strong> neuroblastoma-like areasare in most instances present. These epi<strong>the</strong>lial <strong>and</strong>neuroepi<strong>the</strong>lial elements occur in close relationshipwith each o<strong>the</strong>r <strong>and</strong> with mesenchymal elements, <strong>the</strong>most prominent <strong>of</strong> which are immature cells with ovalor elongated nuclei. The mesenchymal cells may exhibitskeletal muscle differentiation with cross striations(Fig. 2.19c). Foci <strong>of</strong> cartilage, smooth muscle, adiposetissue <strong>and</strong> fibrovascular tissues may also be present. Inspite <strong>of</strong> <strong>the</strong> common occurrence <strong>of</strong> areas having a variety<strong>of</strong> mature tissues, mitotic activity <strong>and</strong> cytologicalfeatures <strong>of</strong> malignancy are demonstrable in <strong>the</strong> undifferentiatedareas <strong>of</strong> both <strong>the</strong> epi<strong>the</strong>lial <strong>and</strong> mesenchymalelements [75].In order to achieve <strong>the</strong> correct diagnosis <strong>of</strong> SNTCSa thorough sampling <strong>of</strong> <strong>the</strong> specimen is required. Inadequatesampling may lead to mistaken diagnoses <strong>of</strong>es<strong>the</strong>sioneuroblastoma, squamous cell carcinoma, undifferentiatedcarcinoma, adenocarcinoma, malignantcraniopharyngioma, malignant mixed tumour <strong>of</strong> salivarygl<strong>and</strong> type, mucoepidermoid carcinoma, adeno-


64 A. Cardesa · L. Alos · A. Franchi2baFig. 2.19. Teratocarcinosarcoma. a CT scan showing massive destructiveinfiltration <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> maxillary sinus on <strong>the</strong>left. With permission [75] b Cystic spaces filled with mucin arepartly covered by benign columnar epi<strong>the</strong>lium <strong>and</strong> surrounded bycimmature blastematous tissue. c Immature gl<strong>and</strong>s surrounded byblastematous elements at <strong>the</strong> lower left; presence <strong>of</strong> immature striatedmuscle, upper right <strong>and</strong> centresquamous carcinoma <strong>and</strong> o<strong>the</strong>rs [105]. In contradistinctionto malignant gonadal teratomas, which arefrequently found in patients at a younger age, sinonasalteratocarcinosarcoma does not contain areas <strong>of</strong> embryonalcarcinoma, choriocarcinoma or germinoma (seminoma),as seen in many germ cell tumours. This makesa germ cell origin <strong>of</strong> SNTCS unlikely [112]. The histogenesis<strong>of</strong> sinonasal teratocarcinosarcoma is debatable.The presence <strong>of</strong> neural tissue in <strong>the</strong>se neoplasms raises<strong>the</strong> possibility that <strong>the</strong> origin could be in some wayrelated to <strong>the</strong> olfactory membrane, or alternatively to<strong>the</strong> sinonasal membrane as a whole, since <strong>the</strong> olfactorymembrane also develops from it [112].Sinonasal teratocarcinosarcomas are locally aggressivetumours, with rapid invasion <strong>of</strong> s<strong>of</strong>t tissues <strong>and</strong>bone, <strong>and</strong> metastasise to regional lymph nodes <strong>and</strong>sites, such as <strong>the</strong> lung. The average survival <strong>of</strong> a patientwith SNTCS is 1.7 years, with 60% <strong>of</strong> <strong>the</strong> patientsnot surviving beyond 3 years. The treatment <strong>of</strong> SNTCSis controversial, but an aggressive initial <strong>the</strong>rapeuticalapproach with a combination <strong>of</strong> surgical resection, radio<strong>the</strong>rapy<strong>and</strong> chemo<strong>the</strong>rapy is usually recommended[112].References1. Abdel-Latif SM, Baheeg SS, Aglan YI, Babin RW, Giltman LI(1987) Chronic atrophic rhinitis with fetor (ozena): a histopathologictreatise. Rhinology 25:117–1202. Abemayor E, Canalis RF, Greenberg P, Wortham DG, Rowl<strong>and</strong>JP, Sun NC (1988) Plasma cell tumors <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck. J Otolaryngol 17:376–3813. Abbondanzo SL, Wenig BM (1995) Non-Hodgkin’s lymphoma<strong>of</strong> <strong>the</strong> sinonasal tract. A clinicopathologic <strong>and</strong> immunophenotypicstudy <strong>of</strong> 124 cases. Cancer 75:1281–12914. Acheson ED, Cowdell RH, Hadfield E, Macbeth RG (1968)Nasal cancer in woodworkers in <strong>the</strong> furniture industry. BrMed J 2:587–5965. Acheson ED, Cowdell RH, Jolles B (1970) Nasal cancer in <strong>the</strong>Northamptonshire boot <strong>and</strong> shoe industry. Br Med J 1:385–3936. Aguilera NS, Kapadia SB, Nalesnik MA, Swerdlow SH (1995)Extramedullary plasmacytoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck: use <strong>of</strong>paraffin sections to assess clonality with in situ hybridization,growth fraction, <strong>and</strong> <strong>the</strong> presence <strong>of</strong> Epstein-Barr virus.Mod Pathol 8:503–5087. Aktas D, Yetiser S, Gerek M, Kurnaz A, Can C, KahramanyolM (1998) Antrochoanal polyps: analysis <strong>of</strong> 16 cases. Rhinology36:81–858. Alobid I, Alos L, Blanch JL, Benitez P, Bernal-Sprekelsen M,Mullol J (2003) Solitary fibrous tumour <strong>of</strong> <strong>the</strong> nasal cavity<strong>and</strong> paranasal sinuses. Acta Otolaryngol 123:71–749. Alos L, Cardesa A, Bombí JA, Mall<strong>of</strong>ré C, Cuchi A, TraserraJ (1996) Myoepi<strong>the</strong>lial tumours <strong>of</strong> salivary gl<strong>and</strong>s: a clinico-


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 65pathologic, immunohistochemical, ultrastructural <strong>and</strong> flowcytometricstudy. Semin Diagn Pathol 13:138–14710. Alos L, Castillo M, Nadal A, Caballero M, Mall<strong>of</strong>re C, PalacinA, Cardesa A (2004) Adenosquamous carcinoma <strong>of</strong> <strong>the</strong>head <strong>and</strong> neck: criteria for diagnosis in a study <strong>of</strong> 12 cases.Histopathology 44:1–1011. Anon JB (2003) Acute bacterial rhinosinusitis in pediatricmedicine: current issues in diagnosis <strong>and</strong> management. PaediatrDrugs 5 [Suppl 1]:25–3312. Apple D, Kreines K (1982) Case report. Cushing’s syndromedue to ectopic ACTH production by a nasal paraganglioma.Am J Med Sci 283:32–3513. Argani P, Perez-Ordonez B, Xiao H, Caruana SM, Huvos AG,Ladanyi M (1998) Olfactory neuroblastoma is not related to<strong>the</strong> Ewing family <strong>of</strong> tumours. Absence <strong>of</strong> EWS/FLI1 gene fusion<strong>and</strong> MIC2 expression. Am J Surg Pathol 22:391–39814. Bailey BJ, Barton S (1975) Olfactory neuroblastoma management<strong>and</strong> prognosis. Arch Otolaryngol 101:1–515. Banerjee AK, Sharma BS, Vashista RK, Kak VK (1992) Intracranialolfactory neuroblastoma: evidence for olfactory epi<strong>the</strong>lialorigin. J Clin Pathol 45:299–30216. Banks ER, Frierson HF, Mills SE et al. (1992) Basaloid squamouscell carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. A clinicopathologic<strong>and</strong> immunohistochemical study <strong>of</strong> 40 cases. Am J SurgPathol 16:939–94617. Barnes L (1986) Intestinal-type adenocarcinoma <strong>of</strong> <strong>the</strong> nasalcavity <strong>and</strong> paranasal sinuses. Am J Surg Pathol 10:192–20218. Barnes L, Bedetti C (1984) Oncocytic Schneiderian papilloma:a reappraisal <strong>of</strong> cylindrical cell papilloma <strong>of</strong> <strong>the</strong> sinonasaltract. Hum Pathol 15:344–35119. Batra K, Chaudhary N, Motwani G, Rai AK (2002) An unusualcase <strong>of</strong> primary nasal tuberculosis with epistaxis <strong>and</strong>epilepsy. Ear Nose Throat J 81:842–84420. Batsakis JG (1970) Mucous gl<strong>and</strong> tumors <strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasalsinuses. Ann Otol Rhinol Laryngol 79:557–56221. Batsakis JG, el-Naggar AK (1992) Rhinoscleroma <strong>and</strong> rhinosporidiosis.Ann Otol Rhinol Laryngol 101:879–88222. Batsakis JG, el-Naggar AK (1996) Cystic fibrosis <strong>and</strong> <strong>the</strong> sinonasaltract. Ann Otol Rhinol Laryngol 105:329–33023. Batsakis JG, Holtz F, Sueper RH (1963) Adenocarcinoma <strong>of</strong><strong>the</strong> nasal <strong>and</strong> paranasal cavities. Arch Otolaryngol 77:625–63324. Batsakis JG, Mackay B, Ordóñez NG (1984) Enteric-type adenocarcinoma<strong>of</strong> <strong>the</strong> nasal cavity. An electron microscopic<strong>and</strong> immunocytochemical study. Cancer 54:855–86025. Batsakis JG, Regezi JA, Solomon AR, Rice DH (1982) The pathology<strong>of</strong> head <strong>and</strong> neck tumors: Mucosal melanomas. <strong>Head</strong><strong>Neck</strong> Surg 4:404–41826. Beatty CW, Pearson BW, Kern EB (1982) Carcinoma <strong>of</strong> <strong>the</strong>nasal septum: experience with 85 cases. Otolaryngol <strong>Head</strong><strong>Neck</strong> Surg 90:90–9427. Begin LR, Rochon L, Frenkel S (1991) Spindle cell myoepi<strong>the</strong>lioma<strong>of</strong> <strong>the</strong> nasal cavity. Am J Surg Pathol 15:184–19028. Benninger MS, Ferguson BJ, Hadley JA, Hamilos DL, JacobsM, Kennedy DW, Lanza DC, Marple BF, OsguthorpeJD, Stankiewicz JA, Anon J, Denneny J, Emanuel I, LevineH (2003) Adult chronic rhinosinusitis: definitions, diagnosis,epidemiology, <strong>and</strong> pathophysiology. Otolaryngol <strong>Head</strong> <strong>Neck</strong>Surg 129:S1–S3229. Ber<strong>the</strong>lsen A, Andersen AP, Jensen S, Hansen HA (1984) Melanomas<strong>of</strong> <strong>the</strong> mucosa in <strong>the</strong> oral cavity <strong>and</strong> <strong>the</strong> upper respiratorypassages. Cancer 54:907–91230. Bosch A, Vallecillo L, Frias Z (1976) Cancer <strong>of</strong> <strong>the</strong> nasal cavity.Cancer 37:1458–146331. Br<strong>and</strong>wein M (2002) Renal cell-like carcinoma <strong>of</strong> <strong>the</strong> sinonasaltract. XXIV International Congress <strong>of</strong> <strong>the</strong> InternationalAcademy <strong>of</strong> <strong>Pathology</strong>. Amsterdam. The Ne<strong>the</strong>rl<strong>and</strong>s 2002.Slide Seminar 9, “Unusual <strong>Head</strong> <strong>and</strong> <strong>Neck</strong> Lesions”. Case 632. Br<strong>and</strong>wein MS, Rothstein A, Lawson W, Bodian C, UrkenML (1997) Sinonasal melanoma. A clinicopathologic study<strong>of</strong> 25 cases <strong>and</strong> literature meta-analysis. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 123:290–29633. Brarsman FA (1980) The median nasal sinus <strong>and</strong> dermoidcyst. Arch Otorhinolaryngol 226:107–11334. Brook I, Frazier EH (2004) Microbiology <strong>of</strong> recurrent acuterhinosinusitis. Laryngoscope 114:129–13135. Buchwald C, Franzmann MB, Jacobsen GK, Lindeberg H(1995) Human papillomavirus (HPV) in sinonasal papillomas:a study <strong>of</strong> 78 cases using in situ hybridization <strong>and</strong> <strong>the</strong>polymerase chain reaction. Laryngoscope 105:66–7136. Buob D, Wacrenier A, Chevalier D, Aubert S, Quinchon JF,Gosselin B, Leroy X (2003) Schwannoma <strong>of</strong> <strong>the</strong> sinonasaltract: a clinicopathologic <strong>and</strong> immunohistochemical study<strong>of</strong> 5 cases. Arch Pathol Lab Med 127:1196–119937. Callender TA, Weber RS, Janjan N, Benjamin R, Zaher M,Wolf P, el-Naggar A (1995) Rhabdomyosarcoma <strong>of</strong> <strong>the</strong> nose<strong>and</strong> paranasal sinuses in adults <strong>and</strong> children. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 112:252–25738. Campo E, Cardesa A, Alos L, Palacin A, Cobarro J, TraserraJ, Montserrat E (1991) Non-Hodgkin’s lymphomas <strong>of</strong> nasalcavity <strong>and</strong> paranasal sinuses. An immunohistochemicalstudy. Am J Clin Pathol 96:184–19039. Cardesa A (1998) Peripheral primitive neuroectodermal tumour,left maxillary sinus. XXII International Congress <strong>of</strong><strong>the</strong> International Academy <strong>of</strong> <strong>Pathology</strong>. Nice. France 1998.Slide Seminar 18, “<strong>Head</strong> <strong>and</strong> <strong>Neck</strong> <strong>Pathology</strong>”. Case 1240. Cardesa A (1998) Sinonasal salivary duct carcinoma metastaticto right cervical lymph nodes. XXII InternationalCongress <strong>of</strong> <strong>the</strong> International Academy <strong>of</strong> <strong>Pathology</strong>. Nice.France 1998. Slide Seminar 18, “<strong>Head</strong> <strong>and</strong> <strong>Neck</strong> <strong>Pathology</strong>”.Case 641. Cardesa A, Alos L (2002) Special tumours <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck region: characterization <strong>of</strong> undifferentiated sinonasaltumours. Histopathology 41 [Suppl 2]:473–47742. Cardesa A, Alos LL, Bombi JA, Palacin A, Traserra J (1991) Relativefrequency <strong>and</strong> diagnoses <strong>of</strong> sinonasal malignant melanoma(SNMM). Lab Invest 64:63A43. Cardesa A, Bombi JA, Alos L (1993) The classification <strong>of</strong> tumours<strong>of</strong> <strong>the</strong> minor salivary gl<strong>and</strong>s. Arquivos de Patologia,Univ. Coimbra, Portugal 25:75–8544. Cardesa A, Pour P, Haas H, Alth<strong>of</strong>f J, Mohr U (1976) Histogenesis<strong>of</strong> tumors from <strong>the</strong> nasal cavities induced by diethylnitrosamine.Cancer 37:346–35545. Carney ME, O’Reilly RC, Sholevar B et al. (1995) Expression<strong>of</strong> <strong>the</strong> human Achaete-scute 1 gene in olfactory neuroblastoma(es<strong>the</strong>sioneuroblastoma). J Neurooncol 26:35–4346. Castro EB, Lewis JS, Strong EW (1973) Plasmacytoma <strong>of</strong> paranasalsinuses <strong>and</strong> nasal cavity. Arch Otolaryngol 97:326–32947. Cecchi F, Buiatti E, Kriebel D, Nastasi L, Santucci M (1980)Adenocarcinoma <strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasal sinuses in shoemakers<strong>and</strong> woodworkers in <strong>the</strong> province <strong>of</strong> Florence, Italy(1963–77). Br J Ind Med 37:222–22548. Cerilli LA, Holst VA, Br<strong>and</strong>wein MS, Stoler MH, Mills SE(2001) Sinonasal undifferentiated carcinoma. Immunohistochemicalpr<strong>of</strong>ile <strong>and</strong> lack <strong>of</strong> EBV association. Am J SurgPathol 25:156–16349. Chan JK (1998) Natural killer cell neoplasms. Anat Pathol3:77–14550. Chan JK, Ng CS, Lau WH, Lo ST (1987) Most nasal/nasopharyngeallymphomas are peripheral T-cell neoplasms. Am JSurg Pathol 11:418–42951. Chaudry MR, Aktar S, Kim DS (1994) Neuroendocrine carcinoma<strong>of</strong> <strong>the</strong> ethmoid sinus. Eur Arch Otorhinolaryngol251:461–46352. Cheung MM, Chan JK, Lau WH et al. (1998) Primary non-Hodgkin’s lymphoma <strong>of</strong> <strong>the</strong> nose <strong>and</strong> nasopharynx: clinicalfeatures, tumor immunophenotype, <strong>and</strong> treatment outcomein 113 patients. J Clin Oncol 16:70–7753. Choi HS, Anderson PJ (1986) Olfactory neuroblastoma: animmuno-electron microscopic study <strong>of</strong> S-100 protein-positivecell. J Neuropathol Exp Neurol 45:576–58754. Christensen WN, Smith RL (1986) Schneiderian papillomas:a clinicopathologic study <strong>of</strong> 67 cases. Hum Pathol 17:393–40055. Cohen MA, Batsakis JG (1968) Oncocytic tumors (oncocytomas)<strong>of</strong> minor salivary gl<strong>and</strong>s. Arch Otolaryngol 88:97–99


66 A. Cardesa · L. Alos · A. Franchi256. Compagno J, Wong RT (1977) Intranasal mixed tumours(pleomorphic adenomas). A clinicopathologic study <strong>of</strong> 40cases. Am J Clin Pathol 68:213–21857. Coup AJ, Hopper IP (1980) Granulomatous lesions in nasalbiopsies. Histopathology 4:293–30858. Cove H (1994) Melanosis, melanocytic hyperplasia, <strong>and</strong>primary malignant melanoma <strong>of</strong> <strong>the</strong> nasal cavity. Cancer44:1424–143359. Cunningham MJ, Brantley S, Barnes L, Schramm VL (1987)Oncocytic Schneiderian papilloma in a young adult: a rarediagnosis. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 97:47–5160. Da-Quan M, Guang-Yan Y (1987) Tumours <strong>of</strong> <strong>the</strong> minor salivarygl<strong>and</strong>s. A clinicopathologic study <strong>of</strong> 243 cases. ActaOtolaryngol (Stockh) 103:325–33161. Davis JM, Weber AL (1980) Pituitary adenoma presenting asa sphenoid sinus lesion. Ann Otol Rhinol Laryngol 89:883–88462. Delgado R, Klimstra D, Albores-Saavedra J (1996) Low gradesalivary duct carcinoma. A distinctive variant with low gradehistology <strong>and</strong> a predominant intraductal growth pattern.Cancer 78:958–96763. Denoyelle F, Ducroz V, Roger G, Garabedian EN (1997) Nasaldermoid sinus cysts in children. Laryngoscope 107:795–80064. Devgan BK, Devgan M, Gross CW (1978) Teratocarcinoma<strong>of</strong> <strong>the</strong> ethmoid sinus: review <strong>of</strong> literature plus a new case report.Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 86:689–69565. Drier JK, Swanson PE, Cherwitz DL, Wick MR (1987) S100protein immunoreactivity in poorly differentiated carcinomas.Immunohistochemical comparison with malignantmelanoma. Arch Pathol Lab Med 111:447–45266. Dun<strong>and</strong> VA, Hammer SM, Rossi R, Poulin M, Albrecht MA,Doweiko JP, DeGirolami PC, Coakley E, Piessens E, WankeCA (1997) Parasitic sinusitis <strong>and</strong> otitis in patients infectedwith human immunodeficiency virus: report <strong>of</strong> five cases<strong>and</strong> review. Clin Infect Dis 25:267–27267. Dykewicz MS (2003) 7. Rhinitis <strong>and</strong> sinusitis. J Allergy ClinImmunol 111:S520–S52968. Eden BV, Debo RF, Larner JM et al. (1994) Es<strong>the</strong>sioneuroblastoma.Long term outcome <strong>and</strong> patterns <strong>of</strong> failure – <strong>the</strong>University <strong>of</strong> Virginia experience. Cancer 73:2556–256269. El-Barbary AE-S, Yassin A, Fouad H, Shennawy ME (1976)Histopathological <strong>and</strong> histochemical studies in atrophic rhinitis.J Laryngol Otol 84:1103–111169a. El-M<strong>of</strong>ty SK, Lu DW (2005) Prevalence <strong>of</strong> high-risk humanpapillomavirus DNA in non-keratinising (cylindrical cell)carcinoma <strong>of</strong> <strong>the</strong> sinonasal tract. A distinct clinicopathologic<strong>and</strong> molecular disease entity. Am J Surg Pathol 29:1367–137270. Elkon D, Hightower SI, Lim ML, Cantrell RW, Constable WC(1979) Es<strong>the</strong>sioneuroblastoma. Cancer 44:1087–109471. Fang SY, Shen CL (1998) Neuropeptide innervation <strong>and</strong> neuroendocrinecells in allergic rhinitis <strong>and</strong> chronic hypertrophicrhinitis. Clin Exp Allergy 28:228–23272. Fellbaum C, Hansmann ML, Lennert K (1989) Malignantlymphomas <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> paranasal sinuses. VirchowsArchiv A Pathol Anat Histopathol 414:399–40573. Ferguson BJ (2000) Mucormycosis <strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasalsinuses. Otolaryngol Clin North Am 33:349–36574. Fern<strong>and</strong>ez PL, Cardesa A, Alos L (1995) Malignant melanoma<strong>of</strong> <strong>the</strong> sinonasal tract. Check Sample. APII. Am J ClinPathol 19:45–5875. Fern<strong>and</strong>ez PL, Cardesa A, Alos L, Pinto J, Traserra J (1995)Sinonasal teratocarcinosarcoma: an unusual neoplasm. PatholRes Pract 191:166–17176. Fern<strong>and</strong>ez PL, Cardesa A, Bombi JA, Palacin A, Traserra J(1993) Malignant sinonasal epi<strong>the</strong>lioid schwannoma. VirchowsArch 423:401–40577. Ferry JA, Sklar J, Zukerberg RL, Harris NL (1991) Nasal lymphoma.A clinicopathologic study with immunophenotypic<strong>and</strong> genotypic analysis. Am J Surg Pathol 15:268–27978. Franchi A, Gallo O, Santucci M (1999) Clinical relevance <strong>of</strong><strong>the</strong> histological classification <strong>of</strong> sinonasal intestinal-type adenocarcinomas.Hum Pathol 30:1140–114579. Franchi A, Massi D, Palomba A, Biancalani M, Santucci M(2004) CDX-2, cytokeratin 7 <strong>and</strong> cytokeratin 20 immunohistochemicalexpression in <strong>the</strong> differential diagnosis <strong>of</strong> primaryadenocarcinomas <strong>of</strong> <strong>the</strong> sinonasal tract. Virchows Arch445:63–6780. Franchi A, Moroni M, Massi D, Paglierani M, Santucci M(2002) Sinonasal undifferentiated carcinoma, nasopharyngeal-typeundifferentiated carcinoma, <strong>and</strong> keratinizing <strong>and</strong>nonkeratinizing squamous cell carcinoma express differentcytokeratin patterns. Am J Surg Pathol 26:1597–160481. Franquemont DW, Fechner RE, Mills SE (1991) Histologicclassification <strong>of</strong> sinonasal intestinal-type adenocarcinoma.Am J Surg Pathol 15:368–37582. Franquemont DW, Mills SE (1991) Sinonasal malignantmelanoma. A clinicopathologic <strong>and</strong> immunohistochemicalstudy <strong>of</strong> 14 cases. Am J Clin Pathol 96:689–69783. Freedman HM, DeSanto LW, Devine KD, Weil<strong>and</strong> LH (1973)Malignant melanoma <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> paranasal sinuses.Arch Otolaryngol 97:322–32584. Friedmann I, Osborn DA (1982) Carcinoma <strong>of</strong> <strong>the</strong> surfaceepi<strong>the</strong>lium (including ameloblastoma). In: Friedmann I, OsbornD (eds) <strong>Pathology</strong> <strong>of</strong> granulomas <strong>and</strong> neoplasms <strong>of</strong> <strong>the</strong>nose <strong>and</strong> paranasal sinuses. Churchill Livingstone, Edinburgh,pp 118–13285. Frierson HF Jr, Innes DJ Jr, Mills SE, Wick M (1989) Immunophenotypicanalysis <strong>of</strong> sinonasal non-Hodgkin’s lymphomas.Human Pathol 20:636–64286. Frierson HF, Mills SE, Fechner RE, Taxy JB, Levine PA(1986) Sinonasal undifferentiated carcinoma. An aggressiveneoplasm derived from Schneiderian epi<strong>the</strong>lium <strong>and</strong> distinctfrom olfactory neuroblastoma. Am J Surg Pathol 10:771–77987. Frierson HF Jr, Ross GW, Mills SE, Frankfurter A (1990) Olfactoryneuroblastoma. Additional immunohistochemicalcharacterization. Am J Surg Pathol 94:547–55388. Frodel JL, Larrabee WF, Raisis J (1989) The nasal dermoid.<strong>Head</strong> <strong>Neck</strong> Surg 101:392–39689. Fu YS, Hoover L, Franklin M, Cheng L, Stoler MH (1992)Human papillomavirus identified by nucleic acid hybridizationin concomitant nasal <strong>and</strong> genital papillomas. Laryngoscope102:1014–101990. Fu YS, Perzin KH (1974) Non-epi<strong>the</strong>lial tumors <strong>of</strong> <strong>the</strong> nasalcavity, paranasal sinuses, <strong>and</strong> nasopharynx: a clinicopathologicstudy. I. General features <strong>and</strong> vascular tumors. Cancer33:1275–128891. Fu YS, Perzin KH (1975) Nonepi<strong>the</strong>lial tumors <strong>of</strong> <strong>the</strong> nasalcavity, paranasal sinuses, <strong>and</strong> nasopharynx: a clinicopathologicstudy. IV. Smooth muscle tumors (leiomyoma, leiomyosarcoma)Cancer 35:1300–130892. Gaal K, Sun NCJ, Hern<strong>and</strong>ez AM, Arber DA (2000) SinonasalNK/T-cell lymphomas in <strong>the</strong> United States. Am J SurgPathol 24:1511–151793. Gadeberg CC, Hjelm-Hansen M, Sögaard H et al. (1984) Malignanttumors <strong>of</strong> <strong>the</strong> paranasal sinuses <strong>and</strong> nasal cavity. Aseries <strong>of</strong> 180 patients. Acta Radiol Oncol 23:181–18794. Gerughty RM, Hennigar GR, Brown FM (1984) Adenosquamouscarcinoma <strong>of</strong> <strong>the</strong> nasal, oral <strong>and</strong> laryngeal cavities. Aclinicopathologic survey <strong>of</strong> ten cases. Cancer 22:1140–115595. Ghamrawi KAE, Glennie JM (1974) The value <strong>of</strong> radio<strong>the</strong>rapyin <strong>the</strong> management <strong>of</strong> malignant melanoma <strong>of</strong> <strong>the</strong> nasalcavity. J Laryngol Otol 88:71–7596. Gnepp DR, Henley J, Weiss S, Heffner D (1996) Desmoid fibromatosis<strong>of</strong> <strong>the</strong> sinonasal tract <strong>and</strong> nasopharynx. A clinicopathologicstudy <strong>of</strong> 25 cases. Cancer 78:2572–257997. Goren AD, Harley N, Eisenbud L, Levin S, Cohen N (1980)Clinical <strong>and</strong> radiobiologic features <strong>of</strong> Thorotrast-inducedcarcinoma <strong>of</strong> <strong>the</strong> maxillary sinus. A case report. Oral SurgOral Med Oral Pathol 49:237–24298. Gosavi DK, Moidekar AT (1978) Chemodectoma <strong>of</strong> <strong>the</strong> nose<strong>and</strong> sphenoid sinus. J Laryngol Otol 92:813–81699. Gouldesbrough DR, Martin-Hirsch DP, Lannigan F (1992)Intranasal malignant melanoma arising in an inverted papilloma.Histopathology 20:523–526100. Guarisco JL, Butcher RB (1990) Congenital cystic teratoma <strong>of</strong><strong>the</strong> maxillary sinus. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 103:1035–1038


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 67101. Gupta A, Seiden AM (2003) Nasal leprosy: case study. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 129:608–610102. Hadfield EH, Macbeth RG (1971) Adenocarcinoma <strong>of</strong> ethmoidsin furniture workers. Ann Otol Rhinol Laryngol80:699–703103. H<strong>and</strong>ler SD, Ward PH (1979) Oncocytoma <strong>of</strong> <strong>the</strong> maxillarysinus. Laryngoscope 69:372–376104. Hanna GS, Ali MH (1987) Verrucous carcinoma <strong>of</strong> <strong>the</strong> nasalseptum. J Laryngol Otol 101:184–187105. Harbo G, Grau C, Bundgaard T, Overgaard M, Elbrond O,Sogaard H, Overgaard J (1997) Cancer <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong>paranasal sinuses. A clinico-pathological study <strong>of</strong> 277 patients.Acta Oncol 36:45–50106. Hardy G (1957) The choanal polyp. Ann Otol Laryngol Rhinol66:306–326107. Harrison DFN (1976) Malignant melanomata arising in <strong>the</strong> nasalmucous membrane. J Laryngol Otol 90:993–1005108. Hasegawa SL, Mentzel T, Fletcher CD (1997) Schwannomas<strong>of</strong> <strong>the</strong> sinonasal tract <strong>and</strong> nasopharynx. Mod Pathol 10:777–784109. Heffner DK (1983) Problems in pediatric otorhinolaryngicpathology. I. Sinonasal <strong>and</strong> nasopharyngeal tumors <strong>and</strong>masses with myxoid features. Int J Pediatr Otorhinolaryngol5:77–91110. Heffner DK (1991) Sinonasal <strong>and</strong> laryngeal salivary gl<strong>and</strong> lesions.In: Ellis GL, Auclair PL, Gnepp DR (eds) Surgical pathology<strong>of</strong> salivary gl<strong>and</strong>s. Saunders, Philadelphia, pp 544–559111. Heffner DK, Gnepp DR (1992) Sinonasal fibrosarcomas, malignantschwannomas, <strong>and</strong> “Triton” tumors. A clinicopathologicstudy <strong>of</strong> 67 cases. Cancer 70:1089–1101112. Heffner DK, Hyams VJ (1984) Teratocarcinosarcoma (malignantteratoma?) <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> paranasal sinuses. Cancer53:2140–215113. Heffner DK, Hyams VJ, Hauck KW, Lingeman C (1982) Lowgradeadenocarcinoma <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> paranasal sinuses.Cancer 50:312–322114. Helliwell TR, Yeoh LH, Stell PM (1986) Anaplastic carcinoma<strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasal sinuses. Light microscopy, immunohistochemistry,<strong>and</strong> clinical correlation. Cancer 58:2038–2045115. Hellquist HB (1996) Nasal polyps update. Histopathology.Allergy Asthma Proc 17:237–242116. Herrmann BW, Sotelo-Avila C, Eisenbeis JF (2003) Pediatricsinonasal rhabdomyosarcoma: three cases <strong>and</strong> a review <strong>of</strong><strong>the</strong> literature. Am J Otolaryngol 24:174–180117. Hillstrom RP, Zarbo RJ, Jacobs JR (1990) Nerve sheath tumors<strong>of</strong> <strong>the</strong> paranasal sinuses: electron microscopy <strong>and</strong>histopathologic diagnosis. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg102:257–263118. Ho KL (1980) Primary meningioma <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong>paranasal sinuses. Cancer 46:1442–1447119. Huang HY, Antonescu CR (2003) Sinonasal smooth musclecell tumors: a clinicopathologic <strong>and</strong> immunohistochemicalanalysis <strong>of</strong> 12 cases with emphasis on <strong>the</strong> low-grade end <strong>of</strong><strong>the</strong> spectrum. Arch Pathol Lab Med 127:297–304120. Hwang HC, Mills SE, Patterson K, Gown AM (1998) Expression<strong>of</strong> <strong>and</strong>rogen receptors in nasopharyngeal angi<strong>of</strong>ibroma:an immunohistochemical study <strong>of</strong> 24 cases. Mod Pathol11:1122–1126121. Hyams VJ (1971) Papillomas <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> paranasalsinuses. A clinicopathologic study <strong>of</strong> 315 cases. Ann OtolRhinol Laryngol 80:192–206122. Hyams VJ, Batsakis JG, Michaels L (1988) Tumors <strong>of</strong> <strong>the</strong> upperrespiratory tract <strong>and</strong> ear. In: Atlas <strong>of</strong> Tumor <strong>Pathology</strong>,2nd series. Fascicle 25. Armed Forces Institute <strong>of</strong> <strong>Pathology</strong>,Washington, D.C.123. Imbus HR, Dyson WL (1987) A review <strong>of</strong> nasal cancer in furnituremanufacturing <strong>and</strong> woodworking in North Carolina,<strong>the</strong> United States, <strong>and</strong> o<strong>the</strong>r countries. J Occup Med 29:734–740124. Ironside P, Mat<strong>the</strong>ws J (1975) Adenocarcinoma <strong>of</strong> <strong>the</strong> nose<strong>and</strong> paranasal sinuses in woodworkers in <strong>the</strong> state <strong>of</strong> Victoria,Australia. Cancer 36:1115–1124125. Jacobsson M, Petruson B, Svendsen P, Ber<strong>the</strong>lsen B (1988)Juvenile nasopharyngeal angi<strong>of</strong>ibroma. A report <strong>of</strong> eighteencases. Acta Otolaryngol 105:132–139126. Jarvi O (1945) Heterotopic tumors with an intestinal mucousmembrane structure in <strong>the</strong> nasal cavity. Acta Otolaryngol33:471–85127. Jeng YM, Sung MT, Fang CL, Huang HY, Mao TL, Cheng W,Hsiao CH (2002) Sinonasal undifferentiated carcinoma <strong>and</strong>nasopharyngeal-type undifferentiated carcinoma. Two clinically,biologically, <strong>and</strong> histopathologically distinct entities.Am J Surg Pathol 26:371–376128. Judd R, Zaki SR, C<strong>of</strong>field LM, Evatt BL (1991) Sinonasal papillomas<strong>and</strong> human papillomavirus: human papillomavirus11 detected in fungiform schneiderian papillomas by insitu hybridization <strong>and</strong> <strong>the</strong> polymerase chain reaction. HumPathol 22:550–556129. Kadish S, Goodman M, Wang CC (1976) Olfactory neuroblastoma.A clinical analysis <strong>of</strong> 17 cases. Cancer 37:1571–1576130. Kagan AR, Nussbaum H, Rao A, Chan P, Gilbert H, Hintz B,Ryoo M, Miles J, Rice D (1981) The management <strong>of</strong> carcinoma<strong>of</strong> <strong>the</strong> nasal vestibule. <strong>Head</strong> <strong>Neck</strong> Surg 4:125–128131. Kahn LB (1974) Es<strong>the</strong>sioneuroblastoma. A light <strong>and</strong> electronmicroscopic study. Hum Pathol 5:364–371132. Kameya T, Shimosato Y, Adachi I, Abe K, Ebihara S, Ono I(1980) Neuroendocrine carcinoma <strong>of</strong> <strong>the</strong> paranasal sinus. Amorphological <strong>and</strong> endocrinological study. Cancer 45:330–339133. Kapadia SB (1985) Hematologic diseases: malignant lymphomas,leukemias, plasma cell dyscrasias, histiocytosis X, <strong>and</strong>reactive lymph node lesions. In: Barnes L (ed) Surgical pathology<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck, vol 2. Dekker, New York134. Kapadia SB, Barnes L, Deutsch M (1981) Non-Hodgkin’slymphoma <strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasal sinuses: a study <strong>of</strong> 17cases. <strong>Head</strong> <strong>Neck</strong> Surg 3:490–499135. Kapadia SB, Barnes L, Pelzman K, Mirani N, Heffner DK, BedettiC (1993) Carcinoma ex oncocytic Schneiderian (cylindricalcell) papilloma. Am J Otolaryngol 14:332–338136. Karma P, Rasanen O, Karja J (1977) Nasal gliomas. A review<strong>and</strong> report <strong>of</strong> two cases. Laryngoscope 87:1169–1179137. Katzenstein AL, Sale SR, Greenberger PA (1983) Pathologicfindings in allergic aspergillus sinusitis. A newly recognizedform <strong>of</strong> sinusitis. Am J Surg Pathol 7:439–443138. Kim ST, Kim CW, Han GC, Park C, Jang IH, Cha HE, ChoiG, Lee HM (2001) Malignant triton tumor <strong>of</strong> <strong>the</strong> nasal cavity.<strong>Head</strong> <strong>Neck</strong> 23:1075–1078139. Kleinsasser O (1985) Terminal tubulus adenocarcinoma <strong>of</strong><strong>the</strong> nasal seromucous gl<strong>and</strong>s. A specific entity. Arch Otorhinolaryngol241:183–193140. Kleinsasser O, Schroeder HG (1988) Adenocarcinoma <strong>of</strong> <strong>the</strong>inner nose after exposure to wood dust. Morphological findings<strong>and</strong> relationships between histopathology <strong>and</strong> clinicalbehavior in 79 cases. Arch Otorhinolaryngol 245:1–15141. Klein-Szanto AJ, Boysen M, Reith A (1987) Keratin <strong>and</strong> involucrinin preneoplastic <strong>and</strong> neoplastic lesions. Distributionin <strong>the</strong> nasal mucosa <strong>of</strong> nickel workers. Arch Pathol Lab Med111:1057–1061142. Klintenberg C, Ol<strong>of</strong>sson J, Hellquist H, Sokjer H (1984) Adenocarcinoma<strong>of</strong> <strong>the</strong> ethmoid sinuses. A review <strong>of</strong> 28 caseswith special reference to wood dust exposure. Cancer54:482–488143. Krespi YP, Kuril<strong>of</strong>f DB, Aner M (1995) Sarcoidosis <strong>of</strong> <strong>the</strong> sinonasaltract: a new staging system. Otolaryngol <strong>Head</strong> <strong>Neck</strong>Surg 112:221–227144. Kuruvilla A, Wenig BM, Humphrey DM, Heffner DK (1990)Leiomyosarcoma <strong>of</strong> <strong>the</strong> sinonasal tract. A clinicopathologicstudy <strong>of</strong> nine cases. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg116:1278–1286145. Kyriakos M (1977) Myospherulosis <strong>of</strong> <strong>the</strong> paranasal sinuses,nose <strong>and</strong> middle ear. A possible iatrogenic disease. Am J ClinPathol 67:118–130146. Larsson LG, Martensson G (1972) Maxillary antral cancers.JAMA 219:342–345147. Lee BJ, Park HJ, Heo SC (2003) Organized hematoma <strong>of</strong> <strong>the</strong>maxillary sinus. Acta Otolaryngol 123:869–872


68 A. Cardesa · L. Alos · A. Franchi2148. Lewis MG, Martin JAM (1967) Malignant melanoma <strong>of</strong> <strong>the</strong> nasalcavity in Ug<strong>and</strong>an Africans: relationship <strong>of</strong> ectopic pigmentation.Cancer 20:1699–1705149. Llombart-Bosch A, Terrier-Lacombe J, Peydro-Olaya A,Contesso G (1989) Peripheral neuroectodermal sarcoma <strong>of</strong>s<strong>of</strong>t tissue (peripheral neuroepi<strong>the</strong>lioma): a pathologic study<strong>of</strong> ten cases with differential diagnosis regarding o<strong>the</strong>r small,round-cell sarcomas. Hum Pathol 20:273–280150. Lloreta J, Serrano S, Corominas JM, Ferrer-Padro E (1995)Polymorphous low-grade adenocarcinoma arising in <strong>the</strong> nasalcavities with an associated undifferentiated carcinoma.Ultrastruct Pathol 19:365–370151. Lloreta-Trull J, Mackay B, Troncoso P, Ribalta-Farres T,Smith T, Khorana S (1992) Neuroendocrine tumors <strong>of</strong> <strong>the</strong>nasal cavity: an ultrastructural study <strong>of</strong> 24 cases. UltrstructPathol 16: 165–175152. Lloyd RV, Ch<strong>and</strong>ler WF, Kovaks K, Ryan N (1986) Ectopicpituitary adenomas with normal anterior pituitary gl<strong>and</strong>s.Am J Surg Pathol 108:546–552153. Lohuis PJ, Lipovsky MM, Hoepelman AI, Hordijk GJ, HuizingEH (1997) Leishmania braziliensis presenting as a granulomatouslesion <strong>of</strong> <strong>the</strong> nasal septum mucosa. J Laryngol Otol111:973–975154. Loree TR, Mullins AP, Spellman J, North JH Jr, Hicks WLJr (1999) <strong>Head</strong> <strong>and</strong> neck mucosal melanoma: a 32-year review.Ear Nose Throat J 78:372–375155. Luce D, Gerin M, Leclerc A, Morcet JF, Brugere J, GoldbergM (1993) Sinonasal cancer <strong>and</strong> occupational exposureto formaldehyde <strong>and</strong> o<strong>the</strong>r substances. Int J Cancer 53:224–231156. Luna MA (1995) Critical commentary to “Sino nasal teratocarcinosarcoma”.Pathol Res Pract 191:172157. Lund V (1982) Malignant melanoma <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong>paranasal sinuses. J Laryngol Otol 96:347–355158. Lund VJ, Milroy CM (1991) Fronto-ethmoidal mucoceles: ahistopathological analysis. J Laryngol Otol 105:921–923159. Mackay B, Luna MA, Butler JJ (1976) Adult neuroblastoma.Electronmicroscopic observations in nine cases. Cancer37:1334–1351160. Majumdar B, Kent S (1983) Malignant neoplasms <strong>of</strong> <strong>the</strong> nose<strong>and</strong> paranasal sinuses. A survey <strong>of</strong> cases treated in a regionalcentre. Clin Otolaryngol 8:97–102161. Makannavar JH, Chavan SS (2001) Rhinosporidiosis. A clinicopathologicalstudy <strong>of</strong> 34 cases. Indian J Pathol Microbiol44:17–21162. Manivel C, Wick MR, Dehner LP (1986) Transitional (cylindric)cell carcinoma with endodermal sinus tumor-like features<strong>of</strong> <strong>the</strong> nasopharynx <strong>and</strong> paranasal sinuses. Clinicopathologic<strong>and</strong> immunohistochemical study <strong>of</strong> two cases.Arch Pathol Lab Med 110:198–202163. Mannan AA, Singh MK, Bahadur S, Hatimota P, SharmaMC (2003) Solitary malignant schwannoma <strong>of</strong> <strong>the</strong> nasal cavity<strong>and</strong> paranasal sinuses: report <strong>of</strong> two rare cases. Ear NoseThroat J 82:634–646164. Marks SC, Upadhyay S, Crane L (1996) Cytomegalovirus sinusitis.A new manifestation <strong>of</strong> AIDS. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 122:789–791165. McDonald TJ, DeRemee RA, Kern EB, Harrison EG Jr (1974)Nasal manifestations <strong>of</strong> Wegener’s granulomatosis. Laryngoscope84:2101–2112166. McKinney CD, Mills SE, Franquemont DW (1995) Sinonasalintestinal-type adenocarcinoma. Immunohistochemicalpr<strong>of</strong>ile <strong>and</strong> comparison with colonic adenocarcinoma. ModPathol 8:421–426167. Mendenhall WM, Stringer SP, Cassisi NJ, Mendenhall NP(1999) Squamous cell carcinoma <strong>of</strong> <strong>the</strong> nasal vestibule. <strong>Head</strong><strong>Neck</strong> 21:385–393168. Mentzel T, Bainbridge TC, Katenkamp D (1997) Solitary fibroustumour: clinicopathological, immunohistochemical,<strong>and</strong> ultrastructural analysis <strong>of</strong> 12 cases arising in s<strong>of</strong>t tissues,nasal cavity <strong>and</strong> nasopharynx, urinary bladder <strong>and</strong> prostate.Virchows Arch 430:445–453169. Mesara BW, Batsakis JG (1966) Gl<strong>and</strong>ular tumors <strong>of</strong> <strong>the</strong> upperrespiratory tract. A clinicopathologic assessment. ArchSurg 92:872–878170. Meyer RD, Gaultier CR, Yamashita JT, Babapour R, PitchonHE, Wolfe PR (1994) Fungal sinusitis in patients with AIDS:report <strong>of</strong> 4 cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Medicine (Baltimore)73:69–78171. Michaels L, Hellquist HB (2001) Ear, nose <strong>and</strong> throat histopathology.Springer, Berlin Heidelberg New York172. Michaels L, Lloyd G, Phelps P (2000) Origin <strong>and</strong> spread <strong>of</strong> allergicfungal disease <strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasal sinuses. ClinOtolaryngol 25:518–525173. Michaels L, Young M (1995) Histogenesis <strong>of</strong> papillomas <strong>of</strong> <strong>the</strong>nose <strong>and</strong> paranasal sinuses. Arch Pathol Lab Med 119:821–826174. Mills SE, Fechner RE, Cantrell RW (1982) Aggressive sinonasallesion resembling normal intestinal mucosa. Am J SurgPathol 6:803–809175. Mills SE, Frierson HF (1985) Olfactory neuroblastoma. Aclinicopathologic study <strong>of</strong> 21 cases. Am J Surg Pathol 9:317–327176. Mills SE, Gaffey MJ, Frierson HF (2000) Tumors <strong>of</strong> <strong>the</strong> upperaerodigestive tract <strong>and</strong> ear. Atlas <strong>of</strong> tumor pathology.Third Series. Fascicle 26. Armed Forces Institute <strong>of</strong> <strong>Pathology</strong>.Washington D.C.177. Min YG, Shin JS, Choi SH, Chi JG, Yoon CJ (1995) Primaryciliary dyskinesia: ultrastructural defects <strong>and</strong> clinicalfeatures. Rhinology 33:189–193178. Moore EJ, Kern EB (2001) Atrophic rhinitis: a review <strong>of</strong> 242cases. Am J Rhinol 15:355–361179. Morita A, Ebersold MJ, Olsen KD, Foote RL, Lewis JE, QuastLM (1993) Es<strong>the</strong>sioneuroblastoma: prognosis <strong>and</strong> management.Neurosurgery 32:706–715180. Mufarrij AA, Busaba NY, Zaytoun GM, Gallo GR, Feiner HD(1990) Primary localized amyloidosis <strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasalsinuses. A case report with immunohistochemical observations<strong>and</strong> a review <strong>of</strong> <strong>the</strong> literature. Am J Surg Pathol14:379–383181. Nakashima T, Kimmelman CP, Snow JB Jr (1984) Structure<strong>of</strong> human fetal <strong>and</strong> adult olfactory neuroepi<strong>the</strong>lium. ArchOtolaryngol 110:641–646182. Nakashima T, Kimmelman CP, Snow JB Jr (1985) Olfactorymarker protein in <strong>the</strong> human olfactory pathway. Arch Otolaryngol111:294–297183. Nakayama M, Wenig BM, Heffner DK (1995) Atypical stromalcells in inflammatory nasal polyps: immunohistochemical<strong>and</strong> ultrastructural analysis in defining histogenesis. Laryngoscope105:127–134184. Natvig K, Larsen TE (1978) Mucocele <strong>of</strong> <strong>the</strong> paranasal sinuses.A retrospective clinical <strong>and</strong> histological study. J LaryngolOtol 92:1075–1082185. Navarrete ML, Quesada P, Pellicer M, Ruiz C (1991) Extramedullarynasal plasmacytoma. J Laryngol Otol 105:41–43186. Ng HK, Poon WS, Poon CY, South JR (1988) Intracranial olfactoryneuroblastoma mimicking carcinoma: report <strong>of</strong> twocases. Histopathology 12:393–403187. Nguyen QA, Gibbs PM, Rice DH (1995) Malignant nasalparaganglioma: a case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 113:157–161188. O’Connor GT Jr, Drake CR, Johns ME, Cail WS, Winn HR,Niskanen E (1985) Treatment <strong>of</strong> advanced es<strong>the</strong>sioneuroblastomawith high-dose chemo<strong>the</strong>rapy <strong>and</strong> autologous bonemarrow transplantation. A case report. Cancer 55:347–349189. Oppenheimer EH, Rosenstein BJ (1979) Differential pathology<strong>of</strong> nasal polyps in cystic fibrosis <strong>and</strong> atopy. Lab Invest40:455–449190. Ordoñez NG, Batsakis JG (1986) Acinic cell carcinoma <strong>of</strong> <strong>the</strong>nasal cavity: electron-optic <strong>and</strong> immunohistochemical observations.J Laryngol Otol 100:345–349191. Ordóñez NG, Mackay B (1993) Neuroendocrine tumors <strong>of</strong><strong>the</strong> nasal cavity. Pathol Annu 28:77–111192. Orvidas LJ, Lewis JE, Olsen KD, Weiner JS (1999) Intranasalverrucous carcinoma; relationship to inverting papilloma<strong>and</strong> human papillomavirus. Laryngoscope 109:371–375193. Osborn DA (1956) Transitional cell growths <strong>of</strong> <strong>the</strong> upper respiratorytract. J Laryngol Otol 70:574–587194. Osborn DA (1970) Nature <strong>and</strong> behavior <strong>of</strong> transitional tumorsin <strong>the</strong> upper respiratory tract. Cancer 25:50–60


Nasal Cavity <strong>and</strong> Paranasal Sinuses Chapter 2 69195. Pai SA, Naresh KN, Masih K, Ramarao C, Borges AM (1998)Teratocarcinosarcoma <strong>of</strong> <strong>the</strong> paranasal sinuses: a clinicopathologic<strong>and</strong> immunohistochemical study. Hum Pathol 29:718–722196. Patterson K, Kapur S, Ch<strong>and</strong>ra RS (1986) “Nasal gliomas”<strong>and</strong> related brain heterotopias: a pathologist’s perspective.Pediatr Pathol 5:353–362197. Patuano E, Carrat X, Drouet Y, Barnabe D, Vincey P, Ber<strong>the</strong>lotB (1993) Mucocutaneous leishmaniasis in otorhinolaryngology.Ann Otolaryngol Chir Cervic<strong>of</strong>ac 110:415–419198. Paugh DR, Sullivan MJ(1990) Myospherulosis <strong>of</strong> <strong>the</strong> paranasalsinuses. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 103:117–119199. Perez-Ordoñez B, Caruana SM, Huvos AG, Shah JP (1998)Small cell neuroendocrine carcinoma <strong>of</strong> <strong>the</strong> <strong>of</strong> <strong>the</strong> nasal cavity<strong>and</strong> paranasal sinuses. Hum Pathol 29:826–832200. Perzin KH, Cantor JO, Johannessen JV (1981) Acinic cell carcinomaarising in <strong>the</strong> nasal cavity: report <strong>of</strong> a case with ultrastructuralobservations. Cancer 47:1818–1822201. Perzin KH, Fu YS (1980) Non-epi<strong>the</strong>lial tumors <strong>of</strong> <strong>the</strong> nasalcavity, paranasal sinuses <strong>and</strong> nasopharynx: a clinico-pathologicstudy. XI. Fibrous histiocytomas. Cancer 45:2616–2626202. Perzin KH, Panyu H, Wechter S (1982) Nonepi<strong>the</strong>lial tumors<strong>of</strong> <strong>the</strong> nasal cavity, paranasal sinuses <strong>and</strong> nasopharynx.A clinicopathologic study. XII. Schwann cell tumors (neurilemomaneur<strong>of</strong>ibroma, malignant schwannoma). Cancer50:2193–2202203. Perzin KH, Pushparaj N (1984) Nonepi<strong>the</strong>lial tumors <strong>of</strong> <strong>the</strong>nasal cavity, paranasal sinuses, <strong>and</strong> nasopharynx. A clinicopathologicstudy. XIII. Meningiomas. Cancer 54:1860–1869204. Phillips PP, Gustafson RO, Facer GW (1990) The clinical behavior<strong>of</strong> inverting papilloma <strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasal sinuses;report <strong>of</strong> 112 cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Laryngoscope100:468–469205. Piscioli F, Aldovini D, Bondi A, Eusebi V (1984) Squamouscell carcinoma with sarcoma-like stroma <strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasalsinuses: report <strong>of</strong> two cases. Histopathology 8:633–639206. Pitman KT, Prokopakis EP, Aydogan B, Segas J, Carrau RL,Snyderman CH,Janecka IP, Hanna E, D’Amico F, JohnsonJT (1999) The role <strong>of</strong> skull base surgery for <strong>the</strong> treatment <strong>of</strong>adenoid cystic carcinoma <strong>of</strong> <strong>the</strong> sinonasal tract. <strong>Head</strong> <strong>Neck</strong>21:402–407207. Prasad ML, Jungbluth AA, Iversen K, Huvos AG, Busam KL(2001) Expression <strong>of</strong> melanocytic differentiation markers inmalignant melanomas <strong>of</strong> <strong>the</strong> oral <strong>and</strong> sinonasal mucosa. AmJ Surg Pathol 25:782–787208. Raychowdhuri RN (1965) Oat cell carcinoma <strong>and</strong> paranasalsinuses. J Laryngol Otol 79:253–255209. Regauer S, Anderhuber W, Richtig E, Schachenreiter J, OttA, Beham A (1998) Primary mucosal melanomas <strong>of</strong> <strong>the</strong> nasalcavity <strong>and</strong> paranasal sinuses. A clinicopathologic analysis <strong>of</strong>14 cases. APMIS 106:403–404210. Rejowski JE, Campanella RS, Block LJ (1982) Small cell carcinoma<strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasal sinuses. Otolaryngol <strong>Head</strong><strong>Neck</strong> Surg 90:516–517211. Ridolfi RL, Liberman PH, Erl<strong>and</strong>son RA, Moore OS (1977)Schneiderian papillomas; a clinicopathologic study <strong>of</strong> 30 cases.Am J Surg Pathol 1:43–53212. Ringertz N (1938) <strong>Pathology</strong> <strong>of</strong> malignant tumours arising in<strong>the</strong> nasal <strong>and</strong> paranasal cavities <strong>and</strong> maxilla. Acta OtolaryngolSuppl 27:31–42213. Robbins KT, Fuller LM, Vlasak M et al. (1985) Primary lymphomas<strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> paranasal sinuses. Cancer56:814–819214. Roberts PF, McCann BG (1985) Eosinophilic angiocentricfibrosis <strong>of</strong> <strong>the</strong> upper respiratory tract: a mucosal variant <strong>of</strong>granuloma faciale? A report <strong>of</strong> three cases. Histopathology9:1217–1225215. Robin PE, Powell DJ (1980) Regional node involvement <strong>and</strong>distant metastases in carcinoma <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong> paranasalsinuses. J Laryngol Otol 94:301–309216. Robin PE, Powell DJ, Stassbie JM (1979) Carcinoma <strong>of</strong> <strong>the</strong>nasal cavity <strong>and</strong> paranasal sinuses: incidence <strong>and</strong> presentation<strong>of</strong> different histological types. Clin Otolaryngol 4:431–456217. Rosai J (1978) The nature <strong>of</strong> myospherulosis <strong>of</strong> <strong>the</strong> upper respiratorytract. Am J Clin Pathol 69:475–481218. Rousch GC (1979) Epidemiology <strong>of</strong> cancer <strong>of</strong> <strong>the</strong> nose <strong>and</strong>paranasal sinuses: current concepts. <strong>Head</strong> <strong>Neck</strong> Surg 2:3–11219. Sadler TW (1985) Langman’s medical embryology, 5th edn.Williams & Wilkins, Baltimore220. Sanchez-Casis G, Devine KD, Weil<strong>and</strong> LH (1971) Nasal adenocarcinomasthat closely simulate colonic carcinomas.Cancer 28:714–729221. Sarkar FH, Visscher DW, Kintanar EB, Zarbo RJ, CrissmanJD (1992) Sinonasal schneiderian papillomas: human papillomavirustyping by polymerase chain reaction. Mod Pathol5:329–332222. Schmid KO, Aubock L, Albegger K (1979) Endocrine-amphicrineenteric carcinoma <strong>of</strong> <strong>the</strong> nasal mucosa. Virchows ArchA Pathol Anat Histol 383:329–343223. Schwartz S, Thiel E (1997) Clinical presentation <strong>of</strong> invasiveaspergillosis. Mycoses 40 [Suppl 2]:21–24224. Seifert G (1991) WHO histological typing <strong>of</strong> salivary gl<strong>and</strong>tumours. Springer, Berlin Heidelberg New York225. Seyer BA, Grist W, Muller S (2002) Aggressive destructivemidfacial lesion from cocaine abuse. Oral Surg Oral MedOral Pathol Oral Radiol Endod 94:465–470226. Shanmugaratnam K (1991) WHO histological typing <strong>of</strong> tumours<strong>of</strong> <strong>the</strong> upper respiratory tract <strong>and</strong> ear. Springer, BerlinHeidelberg New York227. Shanmugaratnam K, Kunaratnam N, Chia KB, Chiang GS,Sinniah R (1983) Teratoid carcinosarcoma <strong>of</strong> <strong>the</strong> paranasalsinuses. <strong>Pathology</strong> 15:413–419228. Shimada K, Kobayashi S, Yamadori I, Ohmori M (1988) Myospherulosisin Japan. A report <strong>of</strong> two cases <strong>and</strong> an immunohistochemicalinvestigation. Am J Surg Pathol 12:427–432229. Silva EG, Butler JJ, Mackay B, Goepfert H (1982) Neuroblastomas<strong>and</strong> neuroendocrine carcinomas <strong>of</strong> <strong>the</strong> nasal cavity. Aproposed new classification. Cancer 50:2388–2405230. Sindwani R, Cohen JT, Pilch BZ, Metson RB (2003) Myospherulosisfollowing sinus surgery: pathological curiosityor important clinical entity? Laryngoscope 113:1123–1127231. Singhal SK, Dass A, Mohan H, Venkataramana Y (2002) Primarynasal tuberculosis. J Otolaryngol 31:60–62232. Siniluoto TM, Luotonen JP, Tikkakoski TA, Leinonen AS,Jokinen KE (1993) Value <strong>of</strong> pre-operative embolization insurgery for nasopharyngeal angi<strong>of</strong>ibroma. J Laryngol Otol107:514–521233. Siu LL, Chan V, Chan JK, Wong KF, Liang R, Kwong YL(2000) Consistent patterns <strong>of</strong> allelic loss in natural killer celllymphoma. Am J Pathol 157:1803–1809234. Skalova A, Cardesa A, Leivo I, Pfaltz M, Ryska A, SimpsonR, Michal M (2003) Sinonasal tubulopapillary low grade adenocarcinoma.Histopathological, immunohistochemical <strong>and</strong>ultrastructural features <strong>of</strong> a poorly recognized entity. VirchowsArch 443:152–158235. Smith CJ, Echevarria R, McLell<strong>and</strong> CA (1974) Pseudosarcomatouschanges in antrochoanal polyps. Arch Otolaryngol99:228–230236. Smith O, Gullane PJ (1987) Inverting papilloma <strong>of</strong> <strong>the</strong> nose:analysis <strong>of</strong> 48 patients. J Otolaryngol 16:154–156237. Sobin LH, Wittekind C (2002) UICC TNM classification <strong>of</strong>malignant tumours, 6th edn. Wiley-Liss, New York238. Sorensen PH, Liu XF, Delattre O et al. (1993) Reverse transcriptasePCR amplification <strong>of</strong> EWS/FLI-1 fusion transcriptsas a diagnostic test for peripheral primitive neuroectodermaltumors <strong>of</strong> childhood. Diagn Mol Pathol 2:147–157239. Spiro RH, Koss LG, Hajdu SI, Strong EW (1973) Tumors <strong>of</strong>minor salivary gl<strong>and</strong> origin: a clinicopathologic study <strong>of</strong> 492cases. Cancer 31:117–129240. Stammberger H (1983) Neue Aspekte zur Genese des invertiertenPapilloms. Laryngol Rhinol Otol (Stuttg) 62:249–255241. Stewart FM, Lazarus HM, Levine PA, Stewart KA, TabbaraIA, Spaulding CA (1989) High-dose chemo<strong>the</strong>rapy <strong>and</strong> autologousmarrow transplantation for es<strong>the</strong>sioneuroblastoma<strong>and</strong> sinonasal undifferentiated carcinoma. Am J Clin Oncol12:217–221


70 A. Cardesa · L. Alos · A. Franchi2242. Stierna P, Carlsoo B (1990) Histopathological observationsin chronic maxillary sinusitis. Acta Otolaryngol 110:450–458243. Sunderman FW Jr, Morgan LG, Andersen A, Ashley D, ForouharFA (1989) Histopathology <strong>of</strong> sinonasal <strong>and</strong> lung cancersin nickel refinery workers. Ann Clin Lab Sci 19:44–50244. Takeshita H, Miwa T, Furukawa M (2002) Osteocartilaginousdifferentiation <strong>of</strong> mucosal melanoma in <strong>the</strong> sinonasalcavity. Ann Otol Rhinol Laryngol 111:1112–1115245. Taxy JB (1997) Squamous carcinoma <strong>of</strong> <strong>the</strong> nasal vestibule.An analysis <strong>of</strong> five cases <strong>and</strong> literature review. Am J ClinPathol 107:698–703246. Taxy JB, Bharani NK, Mills SE, Frierson HF Jr, Gould VE(1986) The spectrum <strong>of</strong> olfactory neural tumors. A light-microscopic,immunohistochemical <strong>and</strong> ultrastructural analysis.Am J Surg Pathol 10:687–705247. Taxy JB, Hidvegi DF (1977) Olfactory neuroblastoma: an ultrastructuralstudy. Cancer 39:131–138248. Thompson LD, Heffner DK (2001) Sinonasal tract eosinophilicangiocentric fibrosis. A report <strong>of</strong> three cases. Am JClin Pathol 115:243–248249. Thompson LD, Miettinen M, Wenig BM (2003) Sinonasaltypehemangiopericytoma: a clinicopathologic <strong>and</strong> immunophenotypicanalysis <strong>of</strong> 104 cases showing perivascular myoiddifferentiation. Am J Surg Pathol 27:737–749250. Thompson LD, Wieneke JA, Miettinen M (2003) Sinonasaltract <strong>and</strong> nasopharyngeal melanomas: a clinicopathologicstudy <strong>of</strong> 115 cases with a proposed staging system. Am J SurgPathol 27:594–611251. Topilko A, Zakrzewski A, Pichard E, Viron A (1984) Ultrastructuralcytochemistry <strong>of</strong> intranuclear dense granules innasopharyngeal angi<strong>of</strong>ibroma. Ultrastruct Pathol 6:221–228252. Torjussen W, Haug FM, Andersen I (1978) Concentration<strong>and</strong> distribution <strong>of</strong> heavy metals in nasal mucosa <strong>of</strong> nickel-exposedworkers <strong>and</strong> <strong>of</strong> controls, studied with atomic absorptionspectrophotometric analysis <strong>and</strong> with Timm’s sulphidesilver method. Acta Otolaryngol 86:449–463253. Torjusen W, Solberg LA, Hogetvit AC (1979) Histopathologicchanges <strong>of</strong> <strong>the</strong> nasal mucosa in nickel workers. A pilot study.Cancer 44:963–974254. Torske KR, Benson GS, Warnock G (2001) Dermoid cyst <strong>of</strong><strong>the</strong> maxillary sinus. Ann Diagn Pathol 5:172–176255. Tran I, Sidrys J, Horton D et al. (1989) Malignant salivarygl<strong>and</strong> tumors <strong>of</strong> <strong>the</strong> paranasal sinuses <strong>and</strong> nasal cavity. TheUCLA experience. Am J Clin Oncol 12:387–392256. Trapp TK, Fu YS, Calcaterra TC (1987) Melanoma <strong>of</strong> <strong>the</strong> nasal<strong>and</strong> paranasal sinus mucosa. <strong>Head</strong> <strong>Neck</strong> Surg 113:1086–1089257. Trojanowski JQ, Lee V, Pillsbury N, Lee S (1982) Neuronalorigin <strong>of</strong> human es<strong>the</strong>sioneuroblastoma demonstrated withanti-neur<strong>of</strong>ilament monoclonal antibodies. N Engl J Med307:159–161258. Tsikoudas A, Martin-Hirsch DP, Woodhead CJ (2001) Primarysinonasal amyloidosis. J Laryngol Otol 115:55–56259. Tufano RP, Mokadam NA, Montone KT Weinstein GS, ChalianAA, Wolf PF, Weber RS (1999) Malignant tumors <strong>of</strong> <strong>the</strong>nose <strong>and</strong> paranasal sinuses: hospital <strong>of</strong> <strong>the</strong> University <strong>of</strong>Pennsylvania experience 1990–1997. Am J Rhinol 13:117–123260. Turc-Carel C, Aurias A, Mugneret F, Lizard S, Sidaner I, VolkC, Thiery JP, Olschwang S, Philip I, Berger MP (1988) Chromosomesin Ewing’s sarcoma. I. An evaluation <strong>of</strong> 85 cases<strong>of</strong> remarkable consistency <strong>of</strong> t(11;22)(q24;q12). Cancer GenetCytogenet 32:229–238261. Walike JW (1973) Anatomy <strong>of</strong> <strong>the</strong> nasal cavities. OtolaryngolClin North Am 6:609–621262. Ward BE, Fechner RE, Mills SE (1990) Carcinoma arising inoncocytic Schneiderian papilloma. Am J Surg Pathol 14:364–369263. Weidner N, Tjoe J (1994) Immunohistochemical pr<strong>of</strong>ile <strong>of</strong>monoclonal antibody O13: antibody that recognizes glycoproteinp30/32MIC2 <strong>and</strong> is useful in diagnosing Ewing’s sarcoma<strong>and</strong> peripheral neuroepi<strong>the</strong>lioma. Am J Surg Pathol18:486–494264. Weiss MD, deFies HO, Taxy JB, Braine H (1983) Primarysmall cell carcinoma <strong>of</strong> <strong>the</strong> paranasal sinuses. Arch Otolaryngol109:341–343265. Weissler MC, Montgomery WW, Montgomery SK, TurnerPA, Joseph MP (1986) Inverted papilloma. Ann Otol RhinolLaryngol 95:215–221266. Wenig BM, Heffner DK (1995) Respiratory epi<strong>the</strong>lial adenomatoidhamartomas <strong>of</strong> <strong>the</strong> sinonasal tract <strong>and</strong> nasopharynx:a clinicopathologic study <strong>of</strong> 31 cases. Ann Otol Rhinol Laryngol104:639–645267. Wenig BM, Hyams VJ, Heffner DK (1988) Nasopharyngealpapillary adenocarcinoma. A clinicopathologic study <strong>of</strong> alow-grade carcinoma. Am J Surg Pathol 12:946–953267a. World Health Organization Classification <strong>of</strong> Tumours (2005)<strong>Pathology</strong> <strong>and</strong> genetics <strong>of</strong> tumours <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck.IARC, Lyon268. Wick MR, Stanley SJ, Swanson PE (1988) Immunohistochemicaldiagnosis <strong>of</strong> sinonasal melanoma, carcinoma, <strong>and</strong>neuroblastoma with monoclonal antibodies HMB-45 <strong>and</strong>antisynaptophysin. Arch Pathol Lab Med 112:616–620269. Wieneke JA, Thompson LD, Wenig BM (1999) Basaloid squamouscell carcinoma <strong>of</strong> <strong>the</strong> sinonasal tract. Cancer 85:841–854270. Wilhelmsson B, Hellquist H, Ol<strong>of</strong>sson J, Klintenberg C (1985)Nasal cuboidal metaplasia with dysplasia. Precursor to adenocarcinomain wood-dust exposed workers? Acta Otolaryngol99:641–648271. Win<strong>the</strong>r B, Gwaltney JM Jr, Mygind N, Hendley JO (1998)Viral-induced rhinitis. Am J Rhinol 12:17–20271a. Wittekind C, Greene FL, Hutter RVP, Klimpfinger M, SobinLH (2004) UICC TNM atlas. Illustrated guide to <strong>the</strong> TNM/pTNM classification <strong>of</strong> malignant tumours. 5 th edn. Springer,Berlin272. Wolff M (1974) Granulomas in nasal mucous membranes followinglocal steroid injections. Am J Clin Pathol 62:775–782273. Woodson GE, Robbins KT, Michaels L (1985) Inverted papilloma.Considerations in treatment. Arch Otolaryngol111:806–811274. Yang YJ, Abraham JL (1997) Undifferentiated carcinomaarising in oncocytic Schneiderian (cylindrical cell) papilloma.J Oral Maxill<strong>of</strong>ac Surg 55:289–294275. Zak FG, Lawson W (1974) The presence <strong>of</strong> melanocytes in <strong>the</strong>nasal cavity. Ann Otol Rhinol Laryngol 83:515–519276. Zarbo RJ, Crissman JD, Venkat H, Weiss MA (1986) Spindlecellcarcinoma <strong>of</strong> <strong>the</strong> upper aerodigestive tract mucosa. Am JSurg Pathol 10:741–753277. Zarbo RJ, Ricci A, Kowalczyk PD, Kartun RW, Knibbs DR(1985) Intranasal dermal analogue tumor (membranous basalcell adenoma). Arch Otolaryngol 111:333–337278. Zerris VA, Annino D, Heilman CB (2002) Nas<strong>of</strong>rontal dermoidsinus cyst: report <strong>of</strong> two cases. Neurosurgery 51:811–814279. Zukerberg LR, Rosenberg AE, R<strong>and</strong>olph G, Pilch BZ, GoodmanML (1991) Solitary fibrous tumor <strong>of</strong> <strong>the</strong> nasal cavity <strong>and</strong>paranasal sinuses. Am J Surg Pathol 15:126–130280. Zur KB, Br<strong>and</strong>wein M, Wang B, Som P Gordon R, Urken ML(2002) Primary description <strong>of</strong> a new entity; renal cell-likecarcinoma <strong>of</strong> <strong>the</strong> nasal cavity. Van Meegeren in <strong>the</strong> house <strong>of</strong>Vermeer. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 128:441–447281. Zurlo JJ, Feuerstein IM, Lebovics R, Lane HC (1992) Sinusitisin HIV-1 infection. Am J Med 93:157–162


Chapter 3Oral Cavity3J.W. EvesonContents3.1 Embryonic Rests <strong>and</strong> Heterotopias . . . . . . . . . . . 723.1.1 Fordyce Granules/Spots . . . . . . . . . . . . . . . . . 723.1.2 Juxtaoral Organ <strong>of</strong> Chievitz . . . . . . . . . . . . . . . 723.2. Vesiculo-Bullous Diseases . . . . . . . . . . . . . . . . 723.2.1 Herpes Simplex Infections . . . . . . . . . . . . . . . . 723.2.2 Chickenpox <strong>and</strong> Herpes Zoster . . . . . . . . . . . . . 733.2.3 H<strong>and</strong>-Foot-<strong>and</strong>-Mouth Disease . . . . . . . . . . . . . 733.2.4 Herpangina . . . . . . . . . . . . . . . . . . . . . . . . . 743.2.5 Pemphigus Vulgaris . . . . . . . . . . . . . . . . . . . . 743.2.6 Pemphigus Vegetans . . . . . . . . . . . . . . . . . . . 743.2.7 Paraneoplastic Pemphigus . . . . . . . . . . . . . . . . 753.2.8 Mucous Membrane Pemphigoid . . . . . . . . . . . . 753.2.9 Dermatitis Herpetiformis . . . . . . . . . . . . . . . . 763.2.10 Linear IgA Disease . . . . . . . . . . . . . . . . . . . . 763.2.11 Ery<strong>the</strong>ma Multiforme . . . . . . . . . . . . . . . . . . 773.3 Ulcerative Lesions . . . . . . . . . . . . . . . . . . . . . 773.3.1 Aphthous Stomatitis(Recurrent Aphthous Ulceration) . . . . . . . . . . . . 773.3.2 Behçet Disease . . . . . . . . . . . . . . . . . . . . . . . 783.3.3 Reiter Disease . . . . . . . . . . . . . . . . . . . . . . . 783.3.4 Median Rhomboid Glossitis . . . . . . . . . . . . . . . 783.3.5 Eosinophilic Ulcer(Traumatic Ulcerative Granulomawith Stromal Eosinophilia) . . . . . . . . . . . . . . . 793.3.6 Acute Necrotising Ulcerative Gingivitis . . . . . . . . 793.3.7 Wegener’s Granulomatosis . . . . . . . . . . . . . . . . 803.3.8 Tuberculosis . . . . . . . . . . . . . . . . . . . . . . . . 813.4 White Lesions . . . . . . . . . . . . . . . . . . . . . . . 813.4.1 C<strong>and</strong>idosis . . . . . . . . . . . . . . . . . . . . . . . . . 813.4.2 Lichen Planus . . . . . . . . . . . . . . . . . . . . . . . 823.4.3 Lupus Ery<strong>the</strong>matosus . . . . . . . . . . . . . . . . . . . 833.4.4 Oral Epi<strong>the</strong>lial Naevi . . . . . . . . . . . . . . . . . . . 843.4.5 Smoker’s Keratosis . . . . . . . . . . . . . . . . . . . . . 843.4.6 Stomatitis Nicotina . . . . . . . . . . . . . . . . . . . . 843.4.7 Hairy Tongue . . . . . . . . . . . . . . . . . . . . . . . 853.4.8 Hairy Leukoplakia . . . . . . . . . . . . . . . . . . . . 853.4.9 Geographic Tongue . . . . . . . . . . . . . . . . . . . . 853.4.10 Frictional Keratosis . . . . . . . . . . . . . . . . . . . . 863.5 Pigmentations . . . . . . . . . . . . . . . . . . . . . . . 863.5.1 Amalgam Tattoo . . . . . . . . . . . . . . . . . . . . . . 863.5.2 Localised Melanotic Pigmentation . . . . . . . . . . . 863.5.2.1 Oral Melanotic Macules . . . . . . . . . . . . . . . . . 863.5.2.2 Melanoacanthoma . . . . . . . . . . . . . . . . . . . . . 873.5.2.3 Pigmented Naevi . . . . . . . . . . . . . . . . . . . . . 873.5.3 Premalignant Oral Melanoses<strong>and</strong> Oral Melanoma . . . . . . . . . . . . . . . . . . . . 873.5.4 Addison Disease . . . . . . . . . . . . . . . . . . . . . . 883.5.5 Peutz Jeghers Syndrome . . . . . . . . . . . . . . . . . 893.5.6 Racial Pigmentation . . . . . . . . . . . . . . . . . . . . 893.5.7 Laugier Hunziker Syndrome . . . . . . . . . . . . . . . 893.5.8 Smoker’s Melanosis . . . . . . . . . . . . . . . . . . . . 893.5.9 Drug-Associated Oral Pigmentation . . . . . . . . . . 903.6 Hyperplastic Lesions . . . . . . . . . . . . . . . . . . . 903.6.1 Fibrous Hyperplasias . . . . . . . . . . . . . . . . . . . 903.6.2 Papillary Hyperplasia . . . . . . . . . . . . . . . . . . . 903.6.3 Generalised Gingival Fibrous Hyperplasia . . . . . . 913.6.4 Crohn’s Disease . . . . . . . . . . . . . . . . . . . . . . 913.6.5 Or<strong>of</strong>acial Granulomatosis . . . . . . . . . . . . . . . . 923.6.6 Chronic Marginal Gingivitis<strong>and</strong> Localised Gingival Fibrous Hyperplasia . . . . . 923.6.7 Peripheral Giant Cell Granuloma(Giant Cell Epulis) . . . . . . . . . . . . . . . . . . . . 933.6.8 Pyogenic Granuloma . . . . . . . . . . . . . . . . . . . 933.6.9 Pulse (Vegetable) Granuloma . . . . . . . . . . . . . . 933.7 Benign Tumours <strong>and</strong> Pseudotumours . . . . . . . . . 943.7.1 Giant Cell Fibroma . . . . . . . . . . . . . . . . . . . . 943.7.2 Lingual Thyroid . . . . . . . . . . . . . . . . . . . . . . 943.7.3 Verruciform Xanthoma . . . . . . . . . . . . . . . . . . . 953.7.4 Haemangiomas . . . . . . . . . . . . . . . . . . . . . . . 953.7.5 Lymphangioma . . . . . . . . . . . . . . . . . . . . . . . 953.7.6 Benign Nerve Sheath Tumours . . . . . . . . . . . . . . 953.7.6.1 Neur<strong>of</strong>ibroma . . . . . . . . . . . . . . . . . . . . . . . 963.7.6.2 Schwannoma . . . . . . . . . . . . . . . . . . . . . . . . 963.7.6.3 Neur<strong>of</strong>ibromatosis . . . . . . . . . . . . . . . . . . . . . 963.7.6.4 Multiple Neuromasin Endocrine Neoplasia Syndrome . . . . . . . . . . . 963.7.7 Granular Cell Tumour(Granular Cell Myoblastoma) . . . . . . . . . . . . . . 963.8 Squamous Cell Carcinoma . . . . . . . . . . . . . . . . 963.8.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 963.8.2 Clinical Features . . . . . . . . . . . . . . . . . . . . . . 973.8.2.1 Buccal Mucosa . . . . . . . . . . . . . . . . . . . . . . . 973.8.2.2 Tongue . . . . . . . . . . . . . . . . . . . . . . . . . . . 973.8.2.3 Floor <strong>of</strong> Mouth . . . . . . . . . . . . . . . . . . . . . . . 973.8.2.4 Gingiva <strong>and</strong> Alveolar Ridge . . . . . . . . . . . . . . . 973.8.2.5 Hard Palate . . . . . . . . . . . . . . . . . . . . . . . . . 983.8.2.6 Retromolar Trigone . . . . . . . . . . . . . . . . . . . . 983.8.3 Staging . . . . . . . . . . . . . . . . . . . . . . . . . . . 98References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98


72 J.W. Eveson33.1 Embryonic Rests<strong>and</strong> Heterotopias3.1.1 Fordyce Granules/SpotsFordyce granules are ectopic sebaceous gl<strong>and</strong>s in <strong>the</strong>oral mucosa [39, 119]. They appear as s<strong>of</strong>t, creamy whiteor yellowish spots or clusters, typically a few millimetresin diameter. They are symmetrically distributed<strong>and</strong> tend to increase in size <strong>and</strong> number with age. Themain site is <strong>the</strong> buccal mucosa, but <strong>the</strong>y may also involve<strong>the</strong> vermilion border <strong>and</strong> labial mucosa, particularly in<strong>the</strong> upper lip. More rarely, <strong>the</strong> tongue, palatoglossal fold,tonsil <strong>and</strong> o<strong>the</strong>r intraoral sites may be affected, <strong>and</strong> <strong>the</strong>condition can <strong>the</strong>n be confused with o<strong>the</strong>r lesions. Microscopyshows typical sebaceous gl<strong>and</strong>s opening directlyonto <strong>the</strong> surface by short, keratinised ducts withno associated hair follicles.3.1.2 Juxtaoral Organ <strong>of</strong> ChievitzChievitz’s organ, or <strong>the</strong> bucco-temporal organ, isthought to be a vestigial neuroepi<strong>the</strong>lial structure. It hasalso been suggested that <strong>the</strong> juxtaoral organ is an anlage<strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>, or arises from Schwann cells thathave undergone squamous metaplasia [132]. It has beendemonstrated in neonates <strong>and</strong> children <strong>and</strong> can persistinto adult life [16]. The organ is usually found between<strong>the</strong> temporalis muscle <strong>and</strong> <strong>the</strong> bucco-temporal fascia orpterygom<strong>and</strong>ibular raphe, <strong>and</strong> is usually present bilaterally.It is seen fortuitously, generally in material takenfrom surgical resections, <strong>and</strong> is important as it can bemisinterpreted as a squamous cell carcinoma. Very rarecases have presented as tumours in <strong>the</strong> infratemporalfossa [83]. It is usually only a few millimetres in size <strong>and</strong>microscopically forms a multilobulated mass <strong>of</strong> discretecell nests that resemble squamous epi<strong>the</strong>lium, but do notshow obvious keratinisation. Occasionally, <strong>the</strong> cells haveclear cytoplasm <strong>and</strong> form duct-like structures that maycontain mucin-negative colloid. The cell nests are associatedwith nerve fibres, particularly at <strong>the</strong> periphery, <strong>and</strong>this may be mistakenly interpreted as a squamous carcinomawith perineural involvement, or sometimes mucoepidermoidcarcinoma <strong>and</strong> thyroid carcinoma [103].The central areas <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lial cell nests are positivefor cytokeratin 19 <strong>and</strong> most cell nests are positive forvimentin <strong>and</strong> weakly positive for epi<strong>the</strong>lial membraneantigen. They are negative for S-100 protein, glial acidicfibrillary protein, <strong>and</strong> neuroendocrine markers suchas chromogranin, synaptophysin <strong>and</strong> neurone-specificenolase [132]. A similar appearance to <strong>the</strong> juxtaoral organhas rarely been described elsewhere in <strong>the</strong> mouth,including intraosseous locations [47].3.2 Vesiculo-Bullous Diseases3.2.1 Herpes Simplex InfectionsHerpes simplex is a common virus that <strong>of</strong>ten causessubclinical infections. It is, however, a cause <strong>of</strong> serious<strong>and</strong> sometimes fatal illnesses in immunocompromisedpatients. In <strong>the</strong> or<strong>of</strong>acial tissues, clinically apparent infectionscan be primary or recurrent. The majority <strong>of</strong>cases <strong>of</strong> oral infections are due to Herpes simplex type1, but an increasing proportion is being attributed toHerpes simplex type 2, which is typically more closelyassociated with genital infections. The virus is transmittedby close contact. Although in <strong>the</strong> past primaryherpes affected children most frequently, in Westernsocieties it is seen increasingly in young <strong>and</strong> middleagedadults.Primary herpes infection (primary herpetic gingivostomatitis)is characterised by widespread vesicular lesions<strong>of</strong> <strong>the</strong> oral mucosa [183]. Any site may be involved,but <strong>the</strong> hard palate <strong>and</strong> <strong>the</strong> dorsum <strong>of</strong> <strong>the</strong> tongue are<strong>the</strong> most common locations. The vesicles quickly ruptureto leave shallow, painful, sharply demarcated ulcersthat are 1–2 mm in diameter <strong>and</strong> have an ery<strong>the</strong>matoushalo. Ulcers frequently coalesce to form more irregularlesions. Gingivitis is a very characteristic feature <strong>of</strong> primaryherpes. The gingivae are swollen <strong>and</strong> <strong>of</strong>ten strikinglyery<strong>the</strong>matous, even in <strong>the</strong> absence <strong>of</strong> frank ulceration.There is <strong>of</strong>ten conspicuous cervical lymphadenopathy,toge<strong>the</strong>r with mild fever <strong>and</strong> malaise. Oral lesionsusually resolve spontaneously within 1–2 weeks.About a third <strong>of</strong> patients infected with Herpes simplex,ei<strong>the</strong>r clinically or sub-clinically, are susceptible to recurrentinfections.It is uncommon for herpetic lesions to be biopsied(Fig 3.1). In <strong>the</strong> early stages <strong>the</strong>re is intercellular oedema<strong>and</strong> ballooning <strong>and</strong> vacuolisation <strong>of</strong> keratinocytesdue to intracellular oedema. This leads to intraepi<strong>the</strong>lialvesiculation. Nuclei become enlarged, <strong>and</strong> occasionallybasophilic or eosinophilic nuclear inclusions witha clear halo ( Lipschutz bodies) can be identified. Cellsmay fuse to form multinucleated epi<strong>the</strong>lial giant cells.The vesiculation is followed rapidly by epi<strong>the</strong>lial necrosis<strong>and</strong> breakdown, leading to ulceration <strong>and</strong> more floridinflammatory infiltration.Herpes simplex virus can persist in a latent form in<strong>the</strong> trigeminal ganglion <strong>and</strong> when re-activated causesrecurrent infections. These are typically seen at <strong>the</strong>mucocutaneous junctions <strong>of</strong> <strong>the</strong> mouth or nasal cavity,<strong>and</strong> involvement <strong>of</strong> <strong>the</strong> lips, <strong>the</strong> most common site,is called herpes labialis. A variety <strong>of</strong> apparently disparatefactors can trigger re-activation, including <strong>the</strong>common cold (“fever blister”), exposure to sunlight,menstruation, stress <strong>and</strong> o<strong>the</strong>rs. There is usually abrief prodromal burning or prickling sensation in <strong>the</strong>


Oral Cavity Chapter 3 73affected area, followed by <strong>the</strong> formation <strong>of</strong> a localisedcluster <strong>of</strong> vesicles. These rapidly break down, ulcerate<strong>and</strong> crust. The lesions usually heal spontaneously in1–2 weeks. Occasionally <strong>the</strong>re may be intraoral recurrences,particularly in <strong>the</strong> hard palate. These may betriggered by local anaes<strong>the</strong>tic injections. Persistentlyrecurrent intraoral herpes, however, should alwaysraise <strong>the</strong> possibility <strong>of</strong> immunosuppression. Atypical<strong>and</strong> <strong>of</strong>ten very severe forms <strong>of</strong> intraoral herpes infectionscan be seen in patients who are immunocompromised[21].3.2.2 Chickenpox<strong>and</strong> Herpes ZosterFig. 3.1. Primary herpetic stomatitis showing intercellular vacuolation<strong>and</strong> multinucleated epi<strong>the</strong>lial cellsChickenpox is a highly contagious infection caused by<strong>the</strong> herpes virus Varicella zoster. It is typically seen inchildren where it causes crops <strong>of</strong> pruritic cutaneousvesicles. It is usually transmitted by direct contact <strong>and</strong>has an incubation period <strong>of</strong> 2–3 weeks. The exan<strong>the</strong>m isfrequently preceded by a slight fever, malaise <strong>and</strong> mildheadache. The cutaneous lesions start as an itchy macularrash, which progressively becomes vesicular <strong>and</strong>pustular before breaking down to form focal crustinglesions. They tend to erupt in crops, but lesions at allstages <strong>of</strong> evolution are frequently present. The back <strong>and</strong>chest are <strong>of</strong>ten <strong>the</strong> first sites <strong>of</strong> involvement, but laterlesions appear on <strong>the</strong> face, neck <strong>and</strong> limbs. They caninvolve <strong>the</strong> nose, ears, conjunctiva <strong>and</strong> genital areas.In <strong>the</strong> mouth <strong>the</strong>y form small, non-specific, scatteredulcers. The symptoms last from a few days to 2 weeks.In many cases <strong>the</strong> virus remains latent in dorsal rootganglia.Herpes zoster (shingles) is due to reactivation <strong>of</strong><strong>the</strong> Varicella zoster virus. In <strong>the</strong> or<strong>of</strong>acial region itis characterised by pain, a vesicular rash <strong>and</strong> stomatitisin <strong>the</strong> related dermatome. Unlike herpes labialis,repeated recurrences <strong>of</strong> zoster are very rare. Occasionally<strong>the</strong>re is an underlying immunodeficiency.Herpes zoster is a hazard in organ transplant patients<strong>and</strong> can be an early complication <strong>of</strong> haematolymphoidneoplasms <strong>and</strong> HIV infections. Herpes zoster usuallyaffects adults <strong>of</strong> middle age or older, but occasionallyinvolves children. The first signs are <strong>of</strong>ten pain, irritationor tenderness in one or more divisions <strong>of</strong> <strong>the</strong>trigeminal nerve. The pain may be severe <strong>and</strong> can bemisinterpreted as toothache, leading to inappropriatedental intervention. Malaise <strong>and</strong> low-grade feverare common constitutional symptoms. There is usuallya strikingly unilateral, vesicular exan<strong>the</strong>m restrictedto <strong>the</strong> affected dermatome. Intraorally, <strong>the</strong>re mayalso be extensive unilateral ulceration in <strong>the</strong> distribution<strong>of</strong> <strong>the</strong> involved nerves. There is usually tender regionallymphadenopathy. The acute phase lasts about7–10 days, but pain may continue until <strong>the</strong> lesions ulcerate<strong>and</strong> crust over, which may take several weeks,especially if <strong>the</strong>re is suppuration <strong>and</strong> subsequent scarring.In <strong>the</strong>se circumstances a significant number <strong>of</strong>patients develop <strong>the</strong> most unpleasant consequence <strong>of</strong>post-herpetic neuralgia.3.2.3 H<strong>and</strong>-Foot-<strong>and</strong>-Mouth DiseaseThis is a common <strong>and</strong> usually mild viral infectionthat <strong>of</strong>ten causes local clusters <strong>of</strong> infections amonggroups <strong>of</strong> young children <strong>and</strong> is characterised by oralulceration <strong>and</strong> a vesicular rash on <strong>the</strong> extremities. Itis caused by a variety <strong>of</strong> strains <strong>of</strong> <strong>the</strong> coxsackie A16virus <strong>and</strong> is highly infectious. Sporadic cases associatedwith Coxsackie A4–7, A9, A10, B1–B3, <strong>and</strong> B5have also been reported. It frequently spreads throughclassrooms, schools <strong>and</strong> local communities in an epidemicmanner. The incubation period is between 3 <strong>and</strong>10 days. It presents clinically as small, scattered oral ulcersthat <strong>of</strong>ten cause few symptoms. Although <strong>the</strong> initiallesions are vesicular, intact blisters are rarely seen.Unlike primary herpes infections, <strong>the</strong> gingivae arerarely affected. It is unusual for regional lymph nodesto be involved except in severe cases <strong>and</strong> constitutionalsymptoms tend to be mild or absent. The cutaneous exan<strong>the</strong>mconsists <strong>of</strong> small vesicles or occasionally largerblisters that form mainly around <strong>the</strong> base <strong>of</strong> fingers ortoes, but may extend to involve any part <strong>of</strong> <strong>the</strong> limb.In some outbreaks, ei<strong>the</strong>r <strong>the</strong> mouth or <strong>the</strong> extremitiesalone may be affected. Although serological investigationscan confirm <strong>the</strong> diagnosis, due to <strong>the</strong> relativelymild <strong>and</strong> transient nature <strong>of</strong> <strong>the</strong> disease, this investigationis rarely undertaken. Typically, <strong>the</strong> condition resolvesspontaneously within a week to 10 days <strong>and</strong> doesnot recur. However, in some epidemics patients havedeveloped severe complications, including interstitialpneumonitis, myocarditis <strong>and</strong> encephalitis, resultingin death [36].


74 J.W. Eveson3.2.4 Herpangina3Herpangina is also caused by a variety <strong>of</strong> group A coxsackieviruses including A1 to 6, 8, 10, <strong>and</strong> 22. O<strong>the</strong>rcauses include coxsackie group B (strains 1–4), echoviruses,<strong>and</strong> o<strong>the</strong>r enteroviruses [161]. It is highly contagious<strong>and</strong> tends to affect young children in <strong>the</strong> summer<strong>and</strong> early autumn period. Like h<strong>and</strong>-foot-<strong>and</strong>-mouthdisease, it rapidly spreads through close-knit communities,such as schools, <strong>and</strong> presents with acute pharyngitis,anorexia <strong>and</strong> dysphagia, with or without cervicallymphadenopathy. Typically, <strong>the</strong> lesions are restricted to<strong>the</strong> s<strong>of</strong>t palate, uvula, anterior pillars <strong>of</strong> <strong>the</strong> fauces <strong>and</strong>palatine tonsils. They consist <strong>of</strong> multiple, small vesiclesthat rapidly rupture to form superficial ulcers, whichmay coalesce. In addition, <strong>the</strong>re is <strong>of</strong>ten more generalisedoropharyngeal ery<strong>the</strong>ma. The condition usuallylasts 1–2 weeks <strong>and</strong> is treated symptomatically.3.2.5 Pemphigus VulgarisFig. 3.2. Pemphigus vulgaris showing suprabasal clefting <strong>and</strong> acantholyticcells in an intraepi<strong>the</strong>lial blisterPemphigus vulgaris is an uncommon, but potentiallylethal, mucocutaneous disorder that occasionally alsoinvolves <strong>the</strong> eyes. It is an autoimmune disease due to circulatingantibodies against <strong>the</strong> intercellular attachments<strong>of</strong> stratified squamous epi<strong>the</strong>lia [163]. The specific targetappears to be desmoglein 3 [2]. However, 50% <strong>of</strong> patientswith pemphigus vulgaris also have autoantibodiesto Dsg1, but cases that are predominantly oral have onlyDsg3 antibodies [70]. Associations have been describedbetween pemphigus vulgaris, myas<strong>the</strong>nia gravis <strong>and</strong>thymoma <strong>and</strong> a variety <strong>of</strong> drugs, including penicillamine,rifampicin <strong>and</strong> captopril. In addition, some casesare associated with internal malignancies, particularly<strong>of</strong> <strong>the</strong> haematolymphoid system, <strong>and</strong> <strong>the</strong> condition is<strong>the</strong>n termed paraneoplastic pemphigus.Pemphigus is more common in Asians <strong>and</strong> AshkenaziJews than o<strong>the</strong>r races <strong>and</strong> most patients are in <strong>the</strong> fourthor fifth decades. The mouth is <strong>the</strong> most common site <strong>of</strong>initial involvement <strong>and</strong> remains <strong>the</strong> only site affected inabout half <strong>of</strong> patients. The oral features are very variable.It is uncommon to find intact vesicles <strong>and</strong> most patientspresent with painful, ragged superficial ulcers <strong>and</strong> areas<strong>of</strong> boggy <strong>and</strong> shredded mucosa. The buccal mucosa, gingiva<strong>and</strong> s<strong>of</strong>t palate are <strong>the</strong> most common sites. In <strong>the</strong>tongue <strong>the</strong> condition may present as deep, non-healingfissures. Fluid from intact or recently ruptured blistersmay contain acantholytic ( Tzanck) cells, although thisis rarely used as a diagnostic measure. The disease maybe relatively mild or even regress, but some cases, particularlythose with extensive cutaneous involvement, maybe fulminant, ei<strong>the</strong>r as a consequence <strong>of</strong> <strong>the</strong> disease itself,or as a complication <strong>of</strong> medical treatment.Microscopy shows suprabasal clefting <strong>of</strong> <strong>the</strong> epi<strong>the</strong>liumdue to loss <strong>of</strong> intercellular attachments <strong>and</strong> acantholysis(Fig 3.2). A single layer <strong>of</strong> keratinocytes may remainattached to <strong>the</strong> corium by <strong>the</strong>ir hemidesmosomes,but <strong>the</strong> cells are separated from each o<strong>the</strong>r laterally t<strong>of</strong>orm a characteristic “tombstone” appearance. The acantholyticcells floating in <strong>the</strong> vesicular fluid are rounded,with condensed cytoplasm surrounding hyperchromaticnuclei. The vesicles may contain acute <strong>and</strong> chronicinflammatory cells, <strong>and</strong> eosinophils may be a conspicuousfeature. In many cases, <strong>the</strong> ro<strong>of</strong> <strong>of</strong> <strong>the</strong> blister is lostduring <strong>the</strong> biopsy procedure due to a positive Nikolskyphenomenon, but a row <strong>of</strong> keratinocytes remains adherentto <strong>the</strong> floor. Direct immun<strong>of</strong>luorescence on frozentissue shows deposits <strong>of</strong> IgG <strong>and</strong> less frequently IgM <strong>and</strong>IgA in <strong>the</strong> intercellular junctions, producing a characteristic“chicken wire” appearance.3.2.6 Pemphigus VegetansPemphigus vegetans is considerably less common in <strong>the</strong>mouth than pemphigus vulgaris [15]. It usually presentsclinically as serpiginous ulcers that are most frequent on<strong>the</strong> dorsum <strong>of</strong> <strong>the</strong> tongue <strong>and</strong> lips [187]. The lingual lesionsclosely resemble those <strong>of</strong> ery<strong>the</strong>ma migrans. Thepapillomatous, proliferative lesions that characterisecutaneous pemphigus vegetans can sometimes be seenat <strong>the</strong> angles <strong>of</strong> <strong>the</strong> mouth. As in pemphigus vulgaris,drugs, particularly ACE inhibitors, have been invokedas possible causative agents in some cases [12, 137].Microscopically, <strong>the</strong> epi<strong>the</strong>lium tends to proliferate<strong>and</strong> become verruciform. Acantholytic cells may not beconspicuous <strong>and</strong> eosinophil microabscesses are said tobe <strong>the</strong> most typical histological feature. However, <strong>the</strong>lesions frequently resemble pyostomatitis vegetans <strong>and</strong>conventional microscopy may not be diagnostic. Thepresence <strong>of</strong> typical skin lesions <strong>of</strong>ten helps in making<strong>the</strong> diagnosis <strong>and</strong> it may be differentiated from pyostomatitisby <strong>the</strong> clinical picture <strong>and</strong> appropriate immunocytochemicalinvestigations [73].


Oral Cavity Chapter 3 753.2.7 Paraneoplastic PemphigusAlthough an occasional association between pemphigus<strong>and</strong> malignancy had been recognised for many years, itwas not until 1990 that paraneoplastic pemphigus wasrecognised as a distinct clinical, histological <strong>and</strong> immunocytochemicalentity [5]. The condition is seenpredominantly in association with B-cell lymphoproliferativedisorders, especially non-Hodgkin lymphoma,chronic lymphocytic leukaemia, Castleman disease,thymoma <strong>and</strong> Waldenström macroglobulinaemia [92].Less commonly it is associated with non-lymphoid neoplasms,including some carcinomas <strong>of</strong> <strong>the</strong> bronchus,breast <strong>and</strong> pancreas. In some cases showing o<strong>the</strong>rwisetypical features <strong>of</strong> <strong>the</strong> disease, no underlying malignancyis found. Paraneoplastic pemphigus is characterisedby <strong>the</strong> following features:■ Painful mucocutaneous vesiculo-bullous eruptions;■ Histopathologic features <strong>of</strong> intraepi<strong>the</strong>lial acantholysis<strong>and</strong> vacuolar interface changes;■ Demonstration <strong>of</strong> intercellular epi<strong>the</strong>lial IgG <strong>and</strong> C3,with or without granular linear deposition <strong>of</strong> complementalong <strong>the</strong> BMZ;■ Presence <strong>of</strong> circulating autoantibodies that bind to<strong>the</strong> surfaces <strong>of</strong> stratified squamous epi<strong>the</strong>lia as wellas simple, columnar <strong>and</strong> transitional epi<strong>the</strong>lia;■ Presence <strong>of</strong> a characteristic complex <strong>of</strong> proteins derivedfrom keratinocytes <strong>and</strong> serum antibodies demonstratedby serum immunoprecipitation. Theseinclude desmoplakins I <strong>and</strong> II, bullous pemphigoidantigen I, envoplakin <strong>and</strong> periplakin [92].In addition, paraneoplastic pemphigus tends to be extremelyrefractory to <strong>the</strong> usual immunosuppressantdrugs used to control pemphigus vulgaris.Paraneoplastic pemphigus is most common between<strong>the</strong> ages <strong>of</strong> 45 <strong>and</strong> 70 years <strong>and</strong> <strong>the</strong>re appears tobe a male predominance. The mouth is almost alwaysinvolved <strong>and</strong> oral lesions present as a painful, intractablestomatitis that extends into <strong>the</strong> oropharynx <strong>and</strong> <strong>of</strong>tenbeyond <strong>the</strong> vermilion borders <strong>of</strong> <strong>the</strong> lips. It causesblisters <strong>and</strong> irregular, ragged ulceration. The buccalmucosa <strong>and</strong> lips are <strong>the</strong> most common sites, but almostanywhere in <strong>the</strong> mouth, oropharynx <strong>and</strong> nasopharynxcan be involved. About two-thirds <strong>of</strong> patients haveconjunctival involvement characterised by frequentlysevere pseudomembranous conjunctivitis <strong>and</strong> symblepharon.Microscopy shows intraepi<strong>the</strong>lial acantholysis withsuprabasal clefting, dyskeratotic keratinocytes, basalcell liquefaction, <strong>and</strong> epi<strong>the</strong>lial inflammatory cellexocytosis [78]. In many cases, however, <strong>the</strong> conditioncannot be distinguished from conventional pemphigus[89]. The overall appearances suggest that <strong>the</strong>reis an overlap between paraneoplastic pemphigus <strong>and</strong>ery<strong>the</strong>ma multiforme. Indirect immun<strong>of</strong>luorescenceon <strong>the</strong> transitional epi<strong>the</strong>lium <strong>of</strong> rat bladder appearsto be a highly specific test for paraneoplastic pemphigus[100].3.2.8 Mucous Membrane PemphigoidMucous membrane pemphigoid is an uncommon, chronicblistering disorder affecting <strong>the</strong> mouth [186]. O<strong>the</strong>rsites <strong>of</strong> involvement include <strong>the</strong> eyes, skin, <strong>and</strong> mucosa<strong>of</strong> <strong>the</strong> nasopharynx, anogenital region, oesophagus <strong>and</strong>larynx [32]. It has been defined as a group <strong>of</strong> putativeautoimmune, chronic inflammatory, subepi<strong>the</strong>lial blisteringdiseases predominantly affecting mucous membranes<strong>and</strong> characterised by linear deposition <strong>of</strong> IgG,IgA <strong>and</strong> C3 along <strong>the</strong> epi<strong>the</strong>lial basement membrane[33]. It has also been called benign mucous membranepemphigoid <strong>and</strong> cicatricial pemphigoid. However, it canbe a severely disabling condition, <strong>and</strong> rarely causes scarringexcept in <strong>the</strong> eye <strong>and</strong> oesophagus/larynx, so <strong>the</strong>seterms are not appropriate.Mucous membrane pemphigoid is more common inwomen than men <strong>and</strong> most patients are in <strong>the</strong> 40–60-year age range. The mouth is <strong>of</strong>ten <strong>the</strong> first <strong>and</strong> onlysite <strong>of</strong> involvement. Oral lesions can be intact blistersthat may contain clear or sero-sanguinous fluid, ery<strong>the</strong>matouspatches or superficial ulcers. Lesions aremost common on <strong>the</strong> attached gingiva, usually buccally<strong>and</strong> labially, <strong>and</strong> <strong>the</strong> palatal mucosa. Less frequentsites include <strong>the</strong> labial, lingual <strong>and</strong> buccal mucosae.Ocular lesions are characterised by conjunctival inflammation,ulceration <strong>and</strong> symblepharon due to fusion<strong>of</strong> <strong>the</strong> palpebral <strong>and</strong> bulbar conjunctivae. Theremay be severe scarring, entropion <strong>and</strong> blindness. Skinlesions are uncommon <strong>and</strong> usually involve <strong>the</strong> scalp<strong>and</strong> upper torso.Microscopy shows subepi<strong>the</strong>lial blister formationwith clean separation <strong>of</strong> <strong>the</strong> full thickness <strong>of</strong><strong>the</strong> epi<strong>the</strong>lium from <strong>the</strong> underlying connective tissue(Fig. 3.3). There is usually a dense mixed inflammatoryinfiltration <strong>of</strong> <strong>the</strong> corium. Due to <strong>the</strong> stronglypositive Nikolsky phenomenon seen in mucous membranepemphigoid, it is very common to receive a biopsyspecimen where most or all <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium hasbeen lost or completely separated from <strong>the</strong> connectivetissue. The specimen <strong>the</strong>n consists <strong>of</strong> non-specificallyinflamed connective tissue, which lacks <strong>the</strong> surfacefibrinous slough that would be more typical <strong>of</strong> a nonspecificoral ulcer. This type <strong>of</strong> appearance, though notdiagnostic, is highly suggestive <strong>of</strong> mucous membranepemphigoid.Direct immun<strong>of</strong>luorescence on peri-lesional mucosalbiopsies shows continuous deposits <strong>of</strong> IgG, IgA or C3, ei<strong>the</strong>rsingly or in combination, along <strong>the</strong> basement membranezone (BMZ) in about 80% <strong>of</strong> cases. When present,<strong>the</strong>se deposits help to distinguish mucous membrane


76 J.W. Eveson3Fig. 3.3. Mucous membrane pemphigoid showing clean subepi<strong>the</strong>lialblisteringFig. 3.4. Dermatitis herpetiformis showing polymorphonuclearleukocytes at <strong>the</strong> tips <strong>of</strong> <strong>the</strong> papillary coriumpemphigoid from several o<strong>the</strong>r common oral mucosalinflammatory disorders. For example, lichen planusdoes not have linear immunoglobulin deposits, but haslinear <strong>and</strong> shaggy deposits <strong>of</strong> fibrin in <strong>the</strong> BMZ, <strong>and</strong> ery<strong>the</strong>mamultiforme has no linear BMZ deposits. However,<strong>the</strong>se deposits do not distinguish mucous membranepemphigoid from bullous pemphigoid, epidermolysisbullosa aquista or linear IgA bullous dermatosis. Suchdistinctions should be made on <strong>the</strong> basis <strong>of</strong> clinical findings.Several possible target antigens have been identifiedin <strong>the</strong> sera <strong>of</strong> patients with mucous membrane pemphigoid.These include: bullous pemphigoid antigens 1 <strong>and</strong>2; laminins 5 <strong>and</strong> 6; type VII collagen <strong>and</strong> ß4 integrinsubunit [32].3.2.9 Dermatitis HerpetiformisDermatitis herpetiformis is an uncommon, intenselypruritic mucocutaneous disorder related to coeliac diseasethat only occasionally involves <strong>the</strong> mouth [126].Oral lesions present as areas <strong>of</strong> ery<strong>the</strong>ma <strong>and</strong> clusters<strong>of</strong> small, friable vesicles or superficial, painful ulcers.The lesions can involve both keratinised <strong>and</strong> non-keratinisedmucosa <strong>and</strong> head <strong>and</strong> neck cutaneous lesionstend to affect <strong>the</strong> scalp <strong>and</strong> periorbital regions. Dermatitisherpetiformis is seen most frequently in teenagers<strong>and</strong> young adults, particularly males, <strong>and</strong> <strong>the</strong>re is a predilectionin people <strong>of</strong> Anglo-Saxon <strong>and</strong> Sc<strong>and</strong>inavianorigin. There is a strong association between dermatitisherpetiformis <strong>and</strong> gluten-sensitive enteropathy. TheClass I antigen HLA-B8 is found in <strong>the</strong> large majority<strong>of</strong> patients with both dermatitis herpetiformis <strong>and</strong> coeliacdisease, <strong>and</strong> HLA-DR3 is expressed in nearly 95%<strong>of</strong> patients.Clinically, oral dermatitis herpetiformis presents aspatches <strong>of</strong> mucosal ery<strong>the</strong>ma, clusters <strong>of</strong> small vesicles,herpetiform ulcers or more extensive areas <strong>of</strong> non-healingulceration. In conventional gluten-sensitive enteropathyoral ulcers tend to be <strong>of</strong> <strong>the</strong> typical minor aphthousstomatitis type.Microscopically, <strong>the</strong> lesions <strong>of</strong> dermatitis herpetiformisshow polymorphonuclear leukocyte microabscessesin <strong>the</strong> tips <strong>of</strong> <strong>the</strong> papillary corium (Fig. 3.4). Initially,neutrophils predominate, but as <strong>the</strong> microabscessesenlarge eosinophils become more conspicuous. The microabscesseseventually fuse to form visible blisters thatfrequently rupture leaving superficial ulcers. Direct immun<strong>of</strong>luorescenceshows granular deposits <strong>of</strong> IgA in <strong>the</strong>BMZ <strong>of</strong> <strong>the</strong> dermal papillae, in both affected <strong>and</strong> adjacentnormal mucosa.3.2.10 Linear IgA diseaseLinear IgA disease is a ra<strong>the</strong>r poorly defined heterogeneousgroup <strong>of</strong> mucocutaneous blistering disordersthat closely resemble mucous membrane pemphigoidclinically <strong>and</strong> microscopically [160, 191]. Like pemphigoid,<strong>the</strong> eyes may be involved. Linear IgA diseasein adults has been separated from similar conditionsin childhood such as bullous dermatosis <strong>of</strong> childhood<strong>and</strong> childhood cicatricial pemphigoid. Cutaneous linearIgA disease <strong>of</strong> adults has a strong association with ahistory <strong>of</strong> bowel disease. This association is much lessclear in patients with oral lesions. However, patientswith oral linear IgA disease appear to have a higherrisk <strong>of</strong> severe ocular lesions. Some cases <strong>of</strong> oral lesionshave been associated with drugs [51]. The conditionis more common in women than men <strong>and</strong> it usuallypresents as a desquamative gingivitis with, or without,ulceration.Microscopy shows subepi<strong>the</strong>lial vesiculation <strong>and</strong>full thickness blister formation. Direct immun<strong>of</strong>luorescenceshows linear deposition <strong>of</strong> IgA along <strong>the</strong> BMZ<strong>and</strong> a low titre <strong>of</strong> circulating IgA to <strong>the</strong> BMZ. Althoughsmall amounts <strong>of</strong> IgG, IgM <strong>and</strong> C3 may be seen, if <strong>the</strong>se


Oral Cavity Chapter 3 77are present in o<strong>the</strong>r than trace amounts mucous membranepemphigoid is a much more likely diagnosis.Linear IgA disease tends to be refractory to systemicsteroids, but it may respond to dapsone or sulphonomides.3.2.11 Ery<strong>the</strong>ma MultiformeEry<strong>the</strong>ma multiforme is a mucocutaneous inflammatorydisorder, but sometimes <strong>the</strong> mouth is <strong>the</strong> only site<strong>of</strong> involvement [1, 8]. It can be relatively mild or manifestwith fever, malaise <strong>and</strong> extensive skin, mucosal<strong>and</strong> ocular lesions when it is sometimes called StevensJohnson syndrome or ery<strong>the</strong>ma multiforme major. Itis thought to be an immunologically mediated disorder,but in many cases no precipitating factor is found.Triggering agents that have been implicated include infectionswith Herpes simplex virus [7] <strong>and</strong> Mycoplasmapneumoniae [99] <strong>and</strong> a wide range <strong>of</strong> drugs includingsulphonamides, anticonvulsants, non-steroidal antiinflammatorymedications <strong>and</strong> antibiotics. Althoughpatients may suffer a single episode, it is <strong>of</strong>ten recurrent.Ery<strong>the</strong>ma multiforme is usually seen in young adults(20–40 years) <strong>and</strong> is more common in males. Oral lesionsmay be <strong>the</strong> only feature <strong>of</strong> <strong>the</strong> disease or cutaneousinvolvement may follow several attacks <strong>of</strong> oral ulceration.The lips are <strong>the</strong> most frequently involved site<strong>and</strong> typically show swelling <strong>and</strong> extensive haemorrhagiccrusting. Within <strong>the</strong> mouth <strong>the</strong>re are usually diffuseery<strong>the</strong>matous areas <strong>and</strong> superficial ulcers on <strong>the</strong> buccalmucosa, floor <strong>of</strong> <strong>the</strong> mouth, tongue, s<strong>of</strong>t palate <strong>and</strong>fauces. It is uncommon for <strong>the</strong> gingiva to be involved<strong>and</strong> this sometimes helps to distinguish ery<strong>the</strong>ma multiformefrom primary herpetic gingivostomatitis, wheregingival inflammation is a conspicuous feature. The areas<strong>of</strong> mucosa involved frequently break down to formpainful, shallow, irregular ulcers on a background <strong>of</strong>more generalised ery<strong>the</strong>ma. It is unusual to see intactblisters in <strong>the</strong> mouth.The classical cutaneous manifestation <strong>of</strong> ery<strong>the</strong>mamultiforme is <strong>the</strong> development <strong>of</strong> so-called target orbull’s eye lesions. These begin as dark red macules, usually1–3 cm in diameter. They become slightly elevated<strong>and</strong> develop a characteristic bluish centre. These lesionsare seen most frequently on <strong>the</strong> h<strong>and</strong>s <strong>and</strong> lower limbs.In ery<strong>the</strong>ma multiforme major <strong>the</strong>re may be ocular <strong>and</strong>genital involvement toge<strong>the</strong>r with constitutional symptoms.A very severe <strong>and</strong> potentially lethal variant is toxicepidermal necrolysis, when <strong>the</strong>re is widespread cutaneous<strong>and</strong> mucosal involvement with extensive blistering<strong>and</strong> epidermal loss leading to fluid <strong>and</strong> electrolyte loss<strong>and</strong> secondary infection.Microscopy shows variable features <strong>and</strong> early epi<strong>the</strong>lialbreakdown <strong>of</strong> oral lesions frequently masks anycharacteristic features [25]. In <strong>the</strong> early lesions <strong>the</strong>re isapoptosis <strong>and</strong> necrosis <strong>of</strong> keratinocytes, intercellular oedema<strong>and</strong> inflammatory infiltration <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium.This leads to intra- <strong>and</strong> sub-epi<strong>the</strong>lial vesiculation <strong>and</strong>ultimately loss <strong>of</strong> <strong>the</strong> ro<strong>of</strong> <strong>of</strong> <strong>the</strong> blister to form an ulcer.There is lymphohistiocytic <strong>and</strong> polymorphonuclear infiltration<strong>of</strong> <strong>the</strong> superficial corium <strong>and</strong> <strong>the</strong> inflammatoryinfiltrate can extend more deeply, <strong>of</strong>ten in a perivasculardistribution. Patchy deposits <strong>of</strong> C3 <strong>and</strong> IgM maybe found in <strong>the</strong> walls <strong>of</strong> blood vessels, but <strong>the</strong>re is n<strong>of</strong>rank vasculitis <strong>and</strong> <strong>the</strong> immune complex depositionappears to be non-specific.3.3 Ulcerative Lesions3.3.1 Aphthous Stomatitis(Recurrent Aphthous Ulceration)This is <strong>the</strong> most common ulcerative disease <strong>of</strong> <strong>the</strong> oralmucosa <strong>and</strong> can affect as many as 15–20% <strong>of</strong> <strong>the</strong> populationat some time in <strong>the</strong>ir lives [152]. It is characterisedby persistently recurrent, painful oral ulcers [142, 143,193]. The condition usually starts in early childhood<strong>and</strong> typically resolves spontaneously in <strong>the</strong> late teens orearly adult life. When <strong>the</strong> condition develops in olderindividuals, predisposing causes such as haematinicdeficiencies or smoking cessation are more likely to beassociated.There are three main clinical forms <strong>of</strong> <strong>the</strong> condition:minor, major <strong>and</strong> herpetiform ulceration, although aminority <strong>of</strong> patients may show various combinations <strong>of</strong><strong>the</strong>se types. Minor aphthae are by far <strong>the</strong> most commonmanifestation (~85%) <strong>and</strong> are characterised by <strong>the</strong> formation<strong>of</strong> one or several superficial ulcers, usually 2–8 mm in diameter with a yellowish-grey, fibrinous floor<strong>and</strong> an ery<strong>the</strong>matous halo. The ulcers tend to involve<strong>the</strong> non-keratinised mucosa such as <strong>the</strong> lips, buccal mucosa,ventrum <strong>of</strong> <strong>the</strong> tongue <strong>and</strong> floor <strong>of</strong> <strong>the</strong> mouth.They usually heal within 7–10 days by regeneration <strong>of</strong><strong>the</strong> epi<strong>the</strong>lium across <strong>the</strong> floor <strong>of</strong> <strong>the</strong> ulcer, <strong>and</strong> withoutscarring. The ulcers frequently recur at regular intervals,typically <strong>of</strong> 2–3 weeks. Some patients, however,are virtually never ulcer free, as new crops appear beforepre-existing ones have healed. A minority <strong>of</strong> casesare menstruation-related <strong>and</strong> <strong>the</strong> ulcers appear monthlyin <strong>the</strong> premenstrual week. Major aphthae are less common(~10%) <strong>and</strong> ulcers can form on both keratinised<strong>and</strong> non-keratinised mucosae. The ulcers are usuallysingle but can be several centimetres in diameter <strong>and</strong>are penetrating. Hence, healing is by secondary intention<strong>and</strong> characterised by granulation tissue formation<strong>and</strong> scarring. In severe cases <strong>the</strong> scarring following progressiveulceration can be so severe that it causes trismus<strong>and</strong> microstomia. Herpetiform aphthae are uncommon


78 J.W. Eveson3(~5%) <strong>and</strong> are characterised by <strong>the</strong> formation <strong>of</strong> sometimeshundreds <strong>of</strong> small (~2 mm), superficial ulcers thatfrequently coalesce <strong>and</strong> may form on a background <strong>of</strong>more generalised mucosal ery<strong>the</strong>ma [154]. Any oral sitemay be involved, but <strong>the</strong> labial <strong>and</strong> ventral lingual mucosaeare <strong>the</strong> sites <strong>of</strong> predilection.Although most cases <strong>of</strong> recurrent aphthous stomatitisare idiopathic, a minority are caused, or exacerbated,by deficiencies in iron, vitamin B 12 or folate, <strong>and</strong> assuch are potentially curable. Haematinic deficienciesare reported to be twice as common in patients withrecurrent aphthous stomatitis compared with controls.The condition is <strong>of</strong>ten made worse by emotional stress.Occasional cases are said to be related to gastro-intestinalcomplaints such as coeliac disease, Crohn’s disease<strong>and</strong> ulcerative colitis, but some <strong>of</strong> <strong>the</strong> data are conflicting[56, 164]. However, it is likely that in most instancesany associations are secondary to haematinic deficiencies.It is uncommon for recurrent aphthae to be biopsied,except when a major aphtha simulates malignancy [97].Reported early changes include infiltration <strong>of</strong> <strong>the</strong> epi<strong>the</strong>liumby lymphocytes <strong>and</strong> histiocytes <strong>and</strong> focal aggregates<strong>of</strong> lymphocytes in <strong>the</strong> superficial corium. Thisis followed by areas <strong>of</strong> epi<strong>the</strong>lial cell apoptosis, degeneration<strong>and</strong> necrosis [77]. The epi<strong>the</strong>lium is lost <strong>and</strong> <strong>the</strong>subsequent ulcer is covered by a fibrinous slough, heavilyinfiltrated by polymorphonuclear leukocytes. Moredeeply <strong>the</strong>re is a mononuclear cell infiltration <strong>and</strong> perivascularcuffing is an inconsistent feature. The conditionappears to be a T-cell mediated immunological response[62] <strong>and</strong> is thought to be a response to a keratinocyte-associatedantigen that is yet to be identified.3.3.2 Behçet DiseaseThis condition comprises recurrent oral ulceration toge<strong>the</strong>rwith genital ulceration <strong>and</strong> ocular lesions [56,88, 109]. The ocular lesions include uveitis <strong>and</strong> retinalvasculitis. Behçet disease, however, is a multisystemdisorder <strong>and</strong> can show a wide range <strong>of</strong> clinical manifestations.Features <strong>of</strong> more generalised disease includeneurological disorders, arthralgia, <strong>and</strong> vascular, gastrointestinal<strong>and</strong> renal lesions. The disease is uncommon<strong>the</strong> USA <strong>and</strong> UK, but has a much higher prevalence insou<strong>the</strong>ast Asia, Japan <strong>and</strong> <strong>the</strong> eastern Mediterranean region.There is a strong association with <strong>the</strong> presence <strong>of</strong>HLA*B51 [116].The oral lesions are clinically identical to recurrentaphthae [177]. Patients also have genital or perigenitalcutaneous ulcers <strong>and</strong> ery<strong>the</strong>ma nodosum is common.Microscopically, like recurrent aphthae, <strong>the</strong> ulcers associatedwith Behçet disease show essentially non-specificfeatures. It has been suggested that perivascular inflammatoryinfiltration into <strong>the</strong> deeper corium may bemore characteristic <strong>of</strong> Behçet disease, but <strong>the</strong> significance<strong>of</strong> this observation is questionable.3.3.3 Reiter DiseaseReiter disease comprises non-specific urethritis, arthritis<strong>and</strong> conjunctivitis, although <strong>the</strong> conjunctivitis ispresent in less than half <strong>of</strong> cases. It was initially thoughtto be only sexually transmitted, but many cases appearto result from enteric infections by a variety <strong>of</strong> organismsincluding Shigella, Salmonella <strong>and</strong> Campylobactor.The disease is typically seen in young males <strong>and</strong>shows a strong association with HLA-B27 <strong>and</strong> has beenreported in HIV-infected individuals [182]. Patients developpainful mono- or polyarticular arthropathy <strong>and</strong>occasionally <strong>the</strong> temporom<strong>and</strong>ibular joint is involved.Patients can have fever, weight loss <strong>and</strong> CNS involvement,<strong>and</strong> facial nerve palsy has been described. Cutaneous<strong>and</strong> mucosal lesions are relatively common. Theskin lesions include macules, vesicles, <strong>and</strong> pustules on<strong>the</strong> h<strong>and</strong>s <strong>and</strong> feet particularly, <strong>and</strong> plaque-like hyperkeratoticlesions <strong>of</strong> <strong>the</strong> trunk <strong>and</strong> scalp. Oral lesionsconsist <strong>of</strong> circinate white or yellowish lesions surroundingmacular areas that are ery<strong>the</strong>matous or superficiallyulcerated. They resemble circinate balanitis <strong>and</strong> <strong>the</strong> lesions<strong>of</strong> geographical tongue <strong>and</strong> geographical stomatitis[131]. They are painless <strong>and</strong> transient <strong>and</strong> are <strong>the</strong>reforerarely biopsied. Microscopy shows features similarto those seen in geographical tongue with spongiformpustules focally <strong>and</strong> diffusely dispersed in <strong>the</strong> superficialepi<strong>the</strong>lium, but without evidence <strong>of</strong> psoriasiformhyperplasia.3.3.4 Median Rhomboid GlossitisMedian rhomboid glossitis usually presents as a painless,reddened, sharply demarcated area <strong>of</strong> depapillationin <strong>the</strong> centre <strong>of</strong> <strong>the</strong> dorsum <strong>of</strong> <strong>the</strong> tongue anterior to<strong>the</strong> foramen caecum. In some cases <strong>the</strong> area is nodularor grooved. It was originally thought to be due to <strong>the</strong>persistence <strong>of</strong> <strong>the</strong> developmental eminence called <strong>the</strong>tuberculum impar, but now most cases are believed tobe c<strong>and</strong>idal in origin [180, 188]. Predisposing factorsinclude smoking, wearing dentures, diabetes <strong>and</strong> usingsteroid inhalers. Sometimes <strong>the</strong>re is a “kissing lesion” in<strong>the</strong> palate.Microscopy typically shows elongation, branching<strong>and</strong> fusion <strong>of</strong> <strong>the</strong> rete ridges with mild epi<strong>the</strong>lial atypia(Fig. 3.5). There may be spongiform pustules in <strong>the</strong>parakeratinised surface layers <strong>and</strong> evidence <strong>of</strong> c<strong>and</strong>idalhyphae. Sometimes <strong>the</strong> epi<strong>the</strong>lial hyperplasia is floridresulting in a pseudoepi<strong>the</strong>liomatous appearance. Some<strong>of</strong> <strong>the</strong>se lesions have been misinterpreted as squamous


Oral Cavity Chapter 3 79Fig. 3.5. Median rhomboid glossitis showing extensive epi<strong>the</strong>lialhyperplasia <strong>and</strong> fusing <strong>of</strong> rete processesFig. 3.6. Eosinophilic ulcers showing plump histiocytic nuclei<strong>and</strong> eosinophilscell carcinomas, with significantly adverse clinical consequences[129]. Below <strong>the</strong> epi<strong>the</strong>lium <strong>the</strong>re is <strong>of</strong>ten adense, b<strong>and</strong>-like zone <strong>of</strong> hyalinisation that is sometimesmistaken for amyloidosis.The lesion <strong>of</strong>ten responds to antifungal treatment,but almost invariably recurs if <strong>the</strong> patient continues tosmoke. There does not appear to be any premalignantpotential <strong>and</strong> <strong>the</strong> dorsum <strong>of</strong> <strong>the</strong> tongue is a very uncommonsite for oral cancer.3.3.5 Eosinophilic Ulcer(Traumatic Ulcerative Granulomawith Stromal Eosinophilia)So-called eosinophilic ulcers are chronic but selflimitinglesions <strong>of</strong> a traumatic or reactive nature inwhich <strong>the</strong>re is an intense inflammatory infiltrationwith a prominent eosinophilic component [45]. Despite<strong>the</strong>ir designation, about a third <strong>of</strong> <strong>the</strong>se lesionsdo not undergo ulceration. Eosinophilic ulcers areseen most commonly in children <strong>and</strong> young adults<strong>and</strong> are frequently painless. In infants, particularly,<strong>the</strong> condition has been called Riga-Fede disease[46]. Although <strong>the</strong>se ulcers can be seen anywhere in<strong>the</strong> oral mucosa, including <strong>the</strong> gingiva, <strong>the</strong>y are mostcommon on <strong>the</strong> tongue <strong>and</strong> buccal mucosa. About athird <strong>of</strong> patients have a history <strong>of</strong> trauma, particularlya crush injury <strong>of</strong> <strong>the</strong> lingual muscle due to biting [43].The lesions are usually single <strong>and</strong> can be several centimetresin diameter. If left, most heal spontaneouslywithin 2 months, <strong>and</strong> this may be accelerated by incisionalbiopsy. Although recurrence tends to be uncommon,in one report 6 out <strong>of</strong> 15 cases were recurrentor multiple [43]. On <strong>the</strong> basis <strong>of</strong> <strong>the</strong> clinical <strong>and</strong>immunocytochemical features it has been suggestedthat eosinophilic ulcers might be <strong>the</strong> oral equivalent<strong>of</strong> primary cutaneous CD30-positive lymphoproliferativedisorders [54].Microscopically, <strong>the</strong>se lesions can be mistaken formalignancy. There is non-specific ulceration with underlyinginflamed granulation tissue (Fig. 3.6). Thereis an associated dense inflammatory infiltrate that extendsdeeply into <strong>the</strong> underlying muscle. The infiltrateconsists <strong>of</strong> lymphocytes <strong>and</strong> plasma cells, macrophages,polymorphonuclear leukocytes <strong>and</strong> mast cells. Eosinophilsare particularly numerous <strong>and</strong> <strong>the</strong>y may form microabscesses.Macrophages are frequently conspicuous<strong>and</strong> can form sheets <strong>of</strong> cells with poorly demarcated cytoplasm,but large, vesicular nuclei with prominent nucleoli<strong>and</strong> a high mitotic frequency. These macrophages,toge<strong>the</strong>r with damaged muscle cells showing sarcolemmalnuclear degeneration <strong>and</strong> regeneration, can give <strong>the</strong>erroneous impression <strong>of</strong> a lymphoma. In addition, <strong>the</strong>prominent eosinophilic component can lead to a mistakendiagnosis <strong>of</strong> Langerhans cell histiocytosis. Thiserroneous diagnosis is particularly likely in lesions involving<strong>the</strong> gingiva, where <strong>the</strong>re may be associated resorption<strong>of</strong> <strong>the</strong> underlying bone. Appropriate immunocytochemicalcharacterisation should avoid this confusion.3.3.6 Acute NecrotisingUlcerative GingivitisAcute necrotising ulcerative gingivitis (Vincent disease,trench mouth) is a relatively common oral disease. Althoughit is generally accepted that bacteria play a pivotalrole in <strong>the</strong> development <strong>of</strong> <strong>the</strong> disease, a specific causalagent has not been established. In <strong>the</strong> past <strong>the</strong> Gramnegativeanaerobes designated as Treponema vincentii<strong>and</strong> Fusobacterium nucleatum were strongly implicated<strong>and</strong> Treponema denticola [165] <strong>and</strong> Prevotella intermediaare some <strong>of</strong> <strong>the</strong> current c<strong>and</strong>idate organisms.A wide variety <strong>of</strong> factors predispose to <strong>the</strong> development<strong>of</strong> <strong>the</strong> disease. The most important local factorsare cigarette smoking <strong>and</strong> poor oral hygiene. General


80 J.W. Eveson3predisposing factors include emotional stress, malnutrition<strong>and</strong> immunosuppressive disorders. Recently, diabeteshas also been implicated in <strong>the</strong> aetiology [102]. Thecondition is most common in young adult males <strong>and</strong> itstarts with painful, punched out <strong>and</strong> crateriform ulcersdeveloping on <strong>and</strong> permanently destroying <strong>the</strong> tips <strong>of</strong><strong>the</strong> interdental papillae. It can spread to <strong>the</strong> marginalgingiva <strong>and</strong> progress to involve <strong>and</strong> destroy <strong>the</strong> underlyingalveolar bone. Lesions sometimes develop in relationto an operculum overlying a partially erupted third molartooth <strong>and</strong> occasionally <strong>the</strong>y spread into <strong>the</strong> adjacentbuccal mucosa. There is <strong>of</strong>ten severe halitosis <strong>and</strong> tastedisturbances. Regional lymphadenopathy is common,but constitutional symptoms tend to be relatively mild.It is very uncommon to receive a biopsy specimenfrom patients with active disease, but scrapings from <strong>the</strong>base <strong>of</strong> one <strong>of</strong> <strong>the</strong> ulcers typically show numerous polymorphs,fibrin <strong>and</strong> debris, <strong>and</strong> suitable staining revealsfusiform <strong>and</strong> spiral organisms.The acute condition usually responds well to oralmetronidazole or tinidazole.3.3.7 Wegener’s GranulomatosisWegener’s granulomatosis (WG) is an uncommon butdistinctive form <strong>of</strong> vasculitis characterised in its classicalform by necrotising granulomatous inflammation<strong>of</strong> <strong>the</strong> upper <strong>and</strong> lower respiratory tracts <strong>and</strong> segmentalnecrotising glomerulonephritis [76]. It is now recognisedthat a wide variety <strong>of</strong> o<strong>the</strong>r organs <strong>and</strong> tissuesmay be involved. Rarely, a proliferative ra<strong>the</strong>r than adestructive response produces tumefactions [65]. Variants<strong>of</strong> WG include a limited form, which has few extrapulmonarymanifestations, <strong>and</strong> a protracted superficialform, which is characterised by lesions restricted to <strong>the</strong>upper respiratory tract, mucosa <strong>and</strong> skin for a prolongedperiod, although it may eventually progress to renal involvement[58]. Wegener’s granulomatosis limited to<strong>the</strong> respiratory system may respond to antibiotics suchas co-trimoxazole <strong>and</strong> Staphylococcus aureus has beenimplicated as a triggering agent for this disease, but <strong>the</strong>evidence remains equivocal [141].<strong>Head</strong> <strong>and</strong> neck manifestations, particularly in <strong>the</strong> sinonasalcomplex, are common <strong>and</strong> can affect as many as90% <strong>of</strong> patients at presentation [50] (see Chap. 2). Theyinclude severe rhinorrhoea, sinusitis, otitis media <strong>and</strong>destruction <strong>of</strong> <strong>the</strong> nasal septum <strong>and</strong> cartilage to producea saddle-nose deformity. By contrast, oral lesionsare less common <strong>and</strong> affect only about 5% <strong>of</strong> patients[69]. They include oral ulceration, delayed healing <strong>of</strong> extractionwounds, tooth mobility <strong>and</strong> loss <strong>of</strong> teeth. Perforation<strong>of</strong> <strong>the</strong> palate is usually as a direct extension <strong>of</strong> sinonasaldisease. Extraorally, head <strong>and</strong> neck manifestationsinclude swelling <strong>and</strong> desquamation <strong>of</strong> <strong>the</strong> lips, parotidgl<strong>and</strong> enlargement, <strong>and</strong> cranial nerve palsies.Fig. 3.7. Wegener’s granulomatosis <strong>of</strong> gingiva showing intenseinflammation, haemorrhage <strong>and</strong> scattered, small multinucleatedgiant cellsA rare, but particularly characteristic oral feature isso-called strawberry gums, which is considered to bevirtually pathognomonic <strong>of</strong> Wegener’s granulomatosis[111, 124]. There is a localised or generalised proliferativegingivitis with a mottled, purplish-red granular surface,which resembles an over-ripe strawberry. Diseaselocalised to <strong>the</strong> gingiva tends to be fairly low-grade. Involvement<strong>of</strong> <strong>the</strong> underlying bone, however, may cause<strong>the</strong> related teeth to loosen or exfoliate.Microscopy <strong>of</strong> <strong>the</strong> gingiva shows irregular epi<strong>the</strong>lialhyperplasia with downgrowths <strong>of</strong> <strong>the</strong> rete processes into<strong>the</strong> underlying corium. The connective tissue shows vascularlakes <strong>of</strong> extravasated blood <strong>and</strong> haemosiderin-containingmacrophages <strong>and</strong> <strong>the</strong>re are neutrophil <strong>and</strong> eosinophilmicroabscesses, toge<strong>the</strong>r with a more diffusemixed inflammatory infiltration (Fig. 3.7). Small multinucleatedgiant cells are unevenly distributed in <strong>the</strong> lesion,<strong>and</strong> although considered to be characteristic, maybe absent in many levels. Vasculitis is not usually seen,possibly because vessels <strong>of</strong> sufficient size to show thisfeature are rarely present in gingivectomy specimens. Bycontrast, biopsies <strong>of</strong> o<strong>the</strong>r oral lesions rarely show microabscessesor necrosis. Also, <strong>the</strong> granulomatous reactioncharacteristic <strong>of</strong> many o<strong>the</strong>r sites is uncommon [42].Investigations <strong>of</strong> patients with suspected oral WGshould include sinus <strong>and</strong> chest radiographs, full bloodpicture, erythrocyte sedimentation rate, C-reactive protein,autoantibody pr<strong>of</strong>ile (including rheumatoid factor)<strong>and</strong> renal function tests. An important investigationis <strong>the</strong> titre <strong>of</strong> antineutrophil cytoplasmic antibody(ANCA), particularly cytoplasmic or cANCA. cANCA isa useful marker <strong>of</strong> WG <strong>and</strong> is found in up to 100% <strong>of</strong> patientswith widespread, active disease [27], but only 60–70% <strong>of</strong> patients with limited forms <strong>of</strong> <strong>the</strong> disease [63].Although cANCA has a very high specificity for WG,it is rarely found in o<strong>the</strong>r types <strong>of</strong> vasculitis [27]. O<strong>the</strong>rANCA-associated vasculitides include microscopicpolyangiitis <strong>and</strong> Churg-Strauss syndrome [38], but


Oral Cavity Chapter 3 81<strong>the</strong>se do not affect <strong>the</strong> mouth. The titre <strong>of</strong> cANCA maybe related to <strong>the</strong> severity <strong>of</strong> <strong>the</strong> disease <strong>and</strong> <strong>the</strong>refore canbe a useful index <strong>of</strong> prognosis <strong>and</strong> efficacy <strong>of</strong> treatment.However, in patients with limited or protracted superficialforms <strong>of</strong> <strong>the</strong> disease, <strong>the</strong> ANCA may be negativefor months or even years so that o<strong>the</strong>r clinicopathologicalcriteria should not be ignored when making <strong>the</strong> diagnosis.3.3.8 TuberculosisOral tuberculosis is rare, but is important as it is usuallya complication <strong>of</strong> advanced open pulmonary disease[155]. Tuberculosis is becoming increasingly more commonin developed countries. This is partly due to HIVinfections <strong>and</strong> <strong>the</strong> fact that multiple drug-resistant mycobacteriaare becoming widespread.The typical lesion is an ulcer, most commonly on <strong>the</strong>mid-dorsum <strong>of</strong> <strong>the</strong> tongue <strong>and</strong> gingiva, but o<strong>the</strong>r sitesmay be involved [122]. The ulcer usually has underminededges, which may be stellate, <strong>and</strong> a pale granularfloor. Occasionally it presents as a non-specific area<strong>of</strong> ery<strong>the</strong>ma or a chronic fissure [112]. It is painless inits early stages, but may become painful later. There isusually no regional lymph node involvement. The clinicalfeatures are <strong>of</strong>ten entirely non-specific <strong>and</strong> <strong>the</strong> diagnosisis initially suspected when <strong>the</strong> microscopy showsmultiple epi<strong>the</strong>lioid granulomas in <strong>the</strong> corium underlyingan ulcer with undermined margins. The granulomasare usually non-caseating <strong>and</strong> it is unusual to demonstrateMycobacteria, even using auramine <strong>and</strong> rhodaminestaining. The organisms may be detected in <strong>the</strong>sputum (but rarely in <strong>the</strong> oral lesion) <strong>and</strong> chest radiographstypically show advanced disease. In patients whoare immunosuppressed, <strong>the</strong> possibility <strong>of</strong> atypical mycobacterialinfection needs to be considered.3.4 White Lesions3.4.1 C<strong>and</strong>idosisOral infections with c<strong>and</strong>idal organisms are very common.The most frequent organism is C<strong>and</strong>ida albicans,a yeast-like fungus [150]. It can cause acute <strong>and</strong> chronicwhite lesions <strong>and</strong> atrophic, red lesions. C<strong>and</strong>idal sporesare present as commensal organisms in <strong>the</strong> mouths <strong>of</strong>as many as 70% <strong>of</strong> individuals. The infective phase <strong>of</strong><strong>the</strong> organism is characterised by <strong>the</strong> presence <strong>of</strong> hyphaethat can directly invade oral keratinocytes [31]. A widevariety <strong>of</strong> factors predispose to infection by c<strong>and</strong>idal organisms,particularly depressed cellular immunity <strong>and</strong>inhibition <strong>of</strong> <strong>the</strong> normal oral flora by broad spectrumantibiotics.Fig. 3.8. C<strong>and</strong>idal hyphae penetrating <strong>the</strong> superficial layers (PAS/D stain)Thrush, or acute hyperplastic c<strong>and</strong>idosis, is seenmost commonly in neonates whose immune systemsare still developing <strong>and</strong> in debilitated patients at <strong>the</strong> extremes<strong>of</strong> life. It is also a feature <strong>of</strong> patients with xerostomiadue to irradiation, Sjögren syndrome <strong>and</strong> a widevariety <strong>of</strong> medications, particularly <strong>the</strong> tricyclic antidepressants.In addition, it is now increasingly becoming afeature <strong>of</strong> immunosuppressed individuals. O<strong>the</strong>r factorspredisposing to <strong>the</strong> development <strong>of</strong> thrush include irondeficiency anaemia, broad spectrum antibiotics <strong>and</strong> steroidinhalers used for <strong>the</strong> control <strong>of</strong> asthma. It is characterisedclinically by <strong>the</strong> formation <strong>of</strong> s<strong>of</strong>t, creamy-white,friable plaques that can be easily wiped <strong>of</strong>f to leave underlyingery<strong>the</strong>matous areas <strong>of</strong> mucosa. The s<strong>of</strong>t palate<strong>and</strong> areas protected from friction such as <strong>the</strong> vestibularreflections are <strong>the</strong> most common sites.Microscopically, <strong>the</strong> characteristic plaque <strong>of</strong> thrushis due to invasion <strong>of</strong> <strong>the</strong> superficial epi<strong>the</strong>lial layers byc<strong>and</strong>idal hyphae <strong>and</strong> <strong>the</strong> subsequent proliferative epi<strong>the</strong>lialresponse (Fig 3.8) [26]. The surface epi<strong>the</strong>lium isparakeratinised, oedematous <strong>and</strong> infiltrated by numerousneutrophils. C<strong>and</strong>idal hyphae penetrate <strong>the</strong> epi<strong>the</strong>liumvertically <strong>and</strong> extend downwards as far as <strong>the</strong> glycogen-richlayer. The hyphae may be inconspicuous inH&E sections unless <strong>the</strong> microscope condenser is loweredto increase <strong>the</strong>ir refractility, but <strong>the</strong>y can be readilyvisualised with periodic acid Schiff or Grocott’s silverstains. The epi<strong>the</strong>lium may show hyperplastic but attenuatedrete processes <strong>and</strong> <strong>the</strong>re is variable but occasionallyflorid acute inflammation <strong>of</strong> <strong>the</strong> underlying corium.Denture-induced stomatitis is a variant <strong>of</strong> atrophicc<strong>and</strong>idosis. It is typically seen in <strong>the</strong> hard palate beneatha full or partial dental pros<strong>the</strong>sis, particularly one constructedfrom acrylic. There is a sharply demarcatedarea <strong>of</strong> bright red, <strong>of</strong>ten boggy ery<strong>the</strong>ma limited by <strong>the</strong>extent <strong>of</strong> <strong>the</strong> denture. Occasionally <strong>the</strong>re may be a fewflecks <strong>of</strong> thrush, but typically <strong>the</strong>re is no plaque formation.Although sometimes referred to as “denture soremouth” <strong>the</strong> condition rarely causes any symptoms un-


82 J.W. Eveson3less it is associated with angular stomatitis. Microscopyshows intercellular oedema <strong>and</strong> chronic inflammatoryinfiltration <strong>of</strong> <strong>the</strong> corium. C<strong>and</strong>idal organisms maynot be seen in biopsy specimens, as <strong>the</strong> fungus tends toproliferate within <strong>the</strong> microscopic interstices <strong>of</strong> <strong>the</strong> denturematerial.Generalised mucosal ery<strong>the</strong>ma, <strong>of</strong>ten with depapillation<strong>of</strong> <strong>the</strong> filiform lingual papillae, can also be a feature<strong>of</strong> both broad-spectrum antibiotic use <strong>and</strong> HIV infection.C<strong>and</strong>idal lesions may present as persistent, adherent,firm white plaques that may be solitary or multiple,particularly in mucocutaneous c<strong>and</strong>idosis syndromes[30]. In <strong>the</strong> latter, <strong>the</strong> mouth is <strong>of</strong>ten <strong>the</strong> most severelyaffected site. These lesions are referred to as chronichyperplastic c<strong>and</strong>idosis or c<strong>and</strong>idal leukoplakia. Mostpatients with isolated plaques are men <strong>of</strong> middle age orolder <strong>and</strong> <strong>the</strong> majority smoke cigarettes. The most commonsites <strong>of</strong> involvement are <strong>the</strong> dorsum <strong>of</strong> <strong>the</strong> tongue<strong>and</strong> <strong>the</strong> post-commissural buccal mucosa. The plaquesare <strong>of</strong>ten thick with a rough, irregular surface that maybe nodular. In many cases <strong>the</strong> lesion forms a variegatedred <strong>and</strong> white patch producing a speckled appearance.Microscopy shows a parakeratinised surface infiltratedby neutrophils forming spongiform pustules. Theepi<strong>the</strong>lium shows downgrowths <strong>of</strong> blunt or club-shapedrete ridges with thinning <strong>of</strong> <strong>the</strong> suprapapillary areas toproduce a psoriasiform appearance. The BMZ may bethickened <strong>and</strong> prominent <strong>and</strong> <strong>the</strong>re is variable but <strong>of</strong>tensevere inflammation in <strong>the</strong> underlying corium. In somecases <strong>the</strong>re can be conspicuous peri-capillary fibrinousexudation, particularly in <strong>the</strong> papillary corium. C<strong>and</strong>idalhyphae may be remarkably sparse <strong>and</strong> not detectedunless multiple sections <strong>and</strong> special stains are used.Electron microscopy shows that <strong>the</strong> hyphae are intracellularparasites that grow within <strong>the</strong> cytoplasm <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lialcells ra<strong>the</strong>r than along <strong>the</strong> intercellular spaces.3.4.2 Lichen PlanusLichen planus is a relatively common chronic inflammatorymucocutaneous disease that can also involve <strong>the</strong>hair <strong>and</strong> nails [162]. Although <strong>the</strong> majority <strong>of</strong> cases <strong>of</strong>cutaneous involvement resolve spontaneously within2–3 years, oral lesions can be remarkably persistent <strong>and</strong>many cases never resolve. It can give rise to white lesions,atrophic areas or superficial ulcers (erosions). Theaetiology is unknown, but <strong>the</strong> histological appearance,which shows T lymphocytes attacking <strong>the</strong> basal epi<strong>the</strong>lium,suggests an autoimmune mechanism. A wide range<strong>of</strong> drugs can precipitate or exacerbate <strong>the</strong> disease <strong>and</strong> asimilar reaction is seen in graft versus host disease.Middle aged or older people are predominantly affected<strong>and</strong> <strong>the</strong> disease is rare in children <strong>and</strong> young adults.Women account for at least 65% <strong>of</strong> patients. White lesionsare frequently asymptomatic, but in some patientslichen planus can lead to intractable oral ulceration thatmay persist for decades. The lesions have characteristicclinical appearances <strong>and</strong> distribution. The most commonform is striae, which are sharply demarcated <strong>and</strong>form lace-like (reticular) or annular patterns. Thesemay be interspersed with defined, small, elevated papules.The patient may complain that <strong>the</strong>y feel a slight restrictionon opening. Less common types <strong>of</strong> white lesionsare confluent plaques, which some term homogeneouslichen planus. They are usually well demarcated,raised plaques <strong>and</strong> are frequently traversed by intersectinggrooves producing a tessellated appearance. The latterappearance is particularly common on <strong>the</strong> dorsum<strong>of</strong> <strong>the</strong> tongue <strong>and</strong> o<strong>the</strong>r sites in long-st<strong>and</strong>ing disease.Atrophic areas, with redness due to mucosal thinningbut without ulceration are usually combined with areas<strong>of</strong> striation. Erosions are shallow, irregular ulcers usuallycovered by a slightly raised, yellowish, fibrinousslough. Very rarely bullae form.Oral lesions <strong>of</strong> lichen planus are very <strong>of</strong>ten symmetrical,sometimes strikingly so, but may be more prominenton one side than ano<strong>the</strong>r. The most frequently affectedsites are <strong>the</strong> buccal mucosae, particularly posteriorly,but lesions may extend to <strong>the</strong> commissures. Thetongue is <strong>the</strong> next most commonly affected site. The lesionsusually involve <strong>the</strong> lateral areas <strong>of</strong> <strong>the</strong> dorsum bilaterallyor less frequently <strong>the</strong> centre <strong>of</strong> <strong>the</strong> dorsum. Theventrum is a relatively uncommon site. Atrophic lichenplanus <strong>of</strong>ten involves <strong>the</strong> gingiva, but reticular lesionsare relatively uncommon at this site. The lips, sometimesincluding <strong>the</strong> vermilion border, may be involved,but <strong>the</strong> palate is rarely affected; lesions <strong>of</strong> <strong>the</strong> floor <strong>of</strong> <strong>the</strong>mouth are exceptional.Sometimes involvement <strong>of</strong> <strong>the</strong> gingiva may be <strong>the</strong>predominant or only manifestation <strong>of</strong> lichen planus. Assuch, it needs to be distinguished from a variety <strong>of</strong> o<strong>the</strong>rinflammatory gingival conditions. The most commonappearance is gingival atrophy <strong>and</strong> <strong>the</strong> epi<strong>the</strong>lial thinningleads to a shiny, red, smooth appearance. This isknown clinically as desquamative gingivitis. It is importantto appreciate that desquamative gingivitis is a clinical,descriptive term <strong>and</strong> not a diagnosis. Diseases o<strong>the</strong>rthan lichen planus that can produce this appearance includemucous membrane pemphigoid, pemphigus <strong>and</strong>a condition called plasma cell gingivitis that is probablyallergy based. Unlike marginal gingivitis, <strong>the</strong> inflammationcan extend onto <strong>the</strong> alveolar mucosa, but in <strong>the</strong>absence <strong>of</strong> secondary plaque accumulation <strong>the</strong>re is usuallysparing <strong>of</strong> <strong>the</strong> marginal gingiva <strong>and</strong> interdental papillae.The condition may be generalised or only patchilydistributed. Also, for unknown reasons, it is rare on<strong>the</strong> lingual <strong>and</strong> palatal gingiva.Clinically, white lesions show parakeratosis or hyperorthokeratosis,sometimes with a prominent granularcell layer [4]. The keratosis may be patchily distrib-


Oral Cavity Chapter 3 83Fig. 3.9. Lichen planus showing basal cell degeneration <strong>and</strong>Civatte bodiesuted as might be expected in reticular or striated lesions.There is a characteristic b<strong>and</strong>-like lymphohistiocytic infiltratesharply localised to <strong>the</strong> superficial corium. Occasionally,germinal centres form within <strong>the</strong> lymphoidinfiltrate. There is <strong>of</strong>ten conspicuous basal cell damagewith apoptosis, ballooning degeneration due to intracellularoedema <strong>and</strong> <strong>the</strong> formation <strong>of</strong> colloid (Civatte) bodies(Fig. 3.9). Fibrinogen deposition along <strong>the</strong> BMZ issometimes a conspicuous feature <strong>and</strong> <strong>the</strong>re may be pigmentaryincontinence secondary to <strong>the</strong> basal cell liquefaction<strong>and</strong> melanophages in <strong>the</strong> superficial corium. Therete ridge pattern is variable, but <strong>the</strong> saw-tooth patterntypical <strong>of</strong> cutaneous lichen planus is relatively uncommonin oral lesions. In lesions from <strong>the</strong> dorsum <strong>of</strong> <strong>the</strong>tongue in particular, <strong>the</strong> rete processes may be elongatedwith dense inflammatory infiltrates around <strong>the</strong>ir tips.Atrophic lesions show conspicuous thinning <strong>and</strong>flattening <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium, but <strong>the</strong> characteristic b<strong>and</strong>likeinflammatory infiltrate is retained. In ulcerated lesions<strong>the</strong> inflammatory infiltrate contains polymorphonuclearleukocytes <strong>and</strong> plasma cells <strong>and</strong> extends into <strong>the</strong>deeper corium, <strong>of</strong>ten leading to a non-specific appearance.As lichen planus is <strong>of</strong>ten treated with topical steroidsit is not uncommon to find infestation <strong>of</strong> <strong>the</strong> superficialepi<strong>the</strong>lial layers by c<strong>and</strong>idal hyphae. These may or maynot be associated with spongiform pustules. Some <strong>of</strong><strong>the</strong>se lesions may also show reactive cytological atypia.A great variety <strong>of</strong> drugs can cause diseases resembling,or in some cases indistinguishable from, lichenplanus. There may be a history relating <strong>the</strong> onset <strong>of</strong> lesionsto <strong>the</strong> drug administration or exacerbation <strong>of</strong> previouslyquiescent disease [104, 105]. The oral lesions are<strong>of</strong>ten severely ulcerated <strong>and</strong> <strong>the</strong> dorsum <strong>of</strong> <strong>the</strong> tongue<strong>and</strong> palate appear to be sites <strong>of</strong> predilection. In some patients,<strong>the</strong>re may be a lichenoid reaction in mucosa indirect contact with dental amalgam fillings <strong>and</strong> occasionallyeven composite filling material <strong>and</strong> gold restorations.Microscopically, <strong>the</strong>re are no absolute diagnostic criteriadistinguishing lichen planus from lichenoid drugeruptions [104]. It is reported that in lichenoid reactions<strong>the</strong> inflammatory infiltrate is more dense. In addition,it is said to be more likely to show <strong>the</strong> presence <strong>of</strong>plasma cells in <strong>the</strong> infiltrate, particularly in <strong>the</strong> leadingedge, <strong>and</strong> a greater likelihood <strong>of</strong> germinal follicle formation.In addition, <strong>the</strong> deep layer <strong>of</strong> <strong>the</strong> infiltrate is lessdefined <strong>and</strong> perivascular inflammatory infiltration extendinginto <strong>the</strong> deeper corium is seen more frequently.However, many <strong>of</strong> <strong>the</strong>se features may reflect <strong>the</strong> moresevere nature <strong>of</strong> <strong>the</strong> disease <strong>and</strong> be related merely to <strong>the</strong>effects <strong>of</strong> ulceration. It is <strong>the</strong>refore essential to examineareas well away from obvious ulceration when interpreting<strong>the</strong>se biopsies.3.4.3 Lupus Ery<strong>the</strong>matosusLupus ery<strong>the</strong>matosus is an autoimmune disease <strong>of</strong>unknown origin. The two main forms that affect <strong>the</strong>mouth are discoid lupus ery<strong>the</strong>matosus (DLE; chroniccutaneous lupus ery<strong>the</strong>matosus) <strong>and</strong> systemic lupus ery<strong>the</strong>matosus(SLE). Oral lesions are present in over 20%<strong>of</strong> patients with SLE.The clinical features <strong>of</strong> oral DLE closely resemblethose <strong>of</strong> lichen planus [157]. They typically show a centralarea <strong>of</strong> atrophic, ery<strong>the</strong>matous or granular mucosawith a surrounding radiating, striated white halo. Thecentral area occasionally ulcerates. Lesions are mostcommon in <strong>the</strong> centre <strong>of</strong> <strong>the</strong> palate <strong>and</strong> on <strong>the</strong> labialaspect <strong>of</strong> <strong>the</strong> upper lip, but <strong>the</strong>y can be seen elsewherein <strong>the</strong> mouth. Sometimes <strong>the</strong>re are adjacent “kissinglesions” on <strong>the</strong> gingiva opposite labial lesions. The lesions<strong>of</strong> DLE lack <strong>the</strong> symmetrical distribution characteristic<strong>of</strong> oral lichen planus. Patients with SLE may have<strong>the</strong> classical photosensitive butterfly rash in <strong>the</strong> midface<strong>and</strong> show o<strong>the</strong>r evidence <strong>of</strong> a systematised disease. Themucosal lesions may resemble those <strong>of</strong> DLE or show evidence<strong>of</strong> more severe mucositis <strong>and</strong> non-specific ulceration.Shallow linear ulcers running parallel to <strong>the</strong> palatalgingiva are sometimes a striking feature.Microscopy shows many similarities to lichen planus[90, 156]. There is ei<strong>the</strong>r hyperorthokeratosis orhyperparakeratosis. The follicular plugging characteristic<strong>of</strong> cutaneous lesions has been described in oral lesions,but it is an inconsistent <strong>and</strong> frequently poorly definedfeature (Fig. 3.10). The rete processes are hyperplastic<strong>and</strong> can form flame-like downgrowths into <strong>the</strong>


84 J.W. Eveson3Fig. 3.10. Discoid lupus ery<strong>the</strong>matosus showing irregular focalepi<strong>the</strong>lial hyperplasia <strong>and</strong> follicular pluggingunderlying corium. This feature is sometimes so floridthat it produces a pseudoepi<strong>the</strong>liomatous appearance.The slender downgrowths can also show a tendency t<strong>of</strong>use with each o<strong>the</strong>r producing an appearance simulatingan embedding artefact. Dyskeratotic cells arean occasional feature. There is apoptosis <strong>and</strong> liquefactiondegeneration <strong>of</strong> <strong>the</strong> basal cells, sometimes withCivatte body formation. The BMZ may become hyalinised<strong>and</strong> thickened <strong>and</strong> this is sometimes a notable feature.There is a b<strong>and</strong>-like infiltrate <strong>of</strong> lymphohistiocyticcells in <strong>the</strong> superficial corium similar to that seen inlichen planus, but <strong>the</strong> lower border tends to be less welldefined <strong>and</strong> <strong>the</strong> infiltrate <strong>of</strong>ten extends into <strong>the</strong> deepertissues in a perivascular distribution. Reactive folliclesmay form in <strong>the</strong> lymphoid infiltrate. In cases <strong>of</strong>SLE <strong>the</strong>re may be evidence <strong>of</strong> fibrinoid necrosis in vessels.Direct immun<strong>of</strong>luorescence <strong>of</strong> lesional tissue forIgG, IgM or IgA shows a granular deposit in <strong>the</strong> BMZ<strong>of</strong> about 75% <strong>of</strong> cases <strong>of</strong> oral DLE <strong>and</strong> all cases <strong>of</strong> SLE.A lupus b<strong>and</strong> test in clinically normal skin or mucosais diagnostic <strong>of</strong> systemic disease. The presence <strong>of</strong> C3 orfibrinogen in <strong>the</strong> BMZ is not specific <strong>and</strong> is frequentlydemonstrated in lichen planus.3.4.4 Oral Epi<strong>the</strong>lial NaeviNon-pigmented epi<strong>the</strong>lial naevi are rare in <strong>the</strong> oral cavity.They include white sponge naevus, oral epi<strong>the</strong>lialnaevi <strong>and</strong> naevi associated with naevus unius lateris.White sponge naevus is an autosomal dominant inheritedtrait characterised by heaped up, sodden, irregularwhite plaques that can affect any part <strong>of</strong> <strong>the</strong> oralmucosa <strong>and</strong> may involve o<strong>the</strong>r mucosae including nasal,anal <strong>and</strong> vaginal (Cannon’s syndrome). Microscopically,<strong>the</strong>re is acanthosis with vacuolated cells in <strong>the</strong> stratumspinosum producing a basket weave appearance<strong>and</strong> irregular, shaggy parakeratosis. It is associated witha novel mutation in <strong>the</strong> keratin 4 gene [34].Oral epi<strong>the</strong>lial naevus was <strong>the</strong> term given to distinctivewhite plaques involving <strong>the</strong> ventral lingual mucosa<strong>and</strong> floor <strong>of</strong> <strong>the</strong> mouth [37]. These were sharply defined,irregularly butterfly shaped <strong>and</strong> had a uniformly wrinkledsurface. However, a retrospective study <strong>of</strong> lesions <strong>of</strong>this type in <strong>the</strong> floor <strong>of</strong> <strong>the</strong> mouth suggested <strong>the</strong>re was asubstantial risk <strong>of</strong> malignant transformation in this lesion<strong>and</strong> it was renamed sublingual keratosis [94, 139].Since that time <strong>the</strong>re have been no o<strong>the</strong>r studies substantiating<strong>the</strong>se observations <strong>and</strong> <strong>the</strong> status <strong>of</strong> <strong>the</strong>se lesionsneeds to be re-established.Naevus unius lateris typically involves <strong>the</strong> skin withunilateral linear, papillary or verrucous lesions, usuallyalong <strong>the</strong> long axis <strong>of</strong> a limb or across <strong>the</strong> trunk.It has uncommonly been associated with oral lesions[74]. These are typically papillary, wart-like proliferations<strong>and</strong> have been described on <strong>the</strong> lips, tongue, buccalmucosa, palate <strong>and</strong> gingivae, usually on <strong>the</strong> left. Microscopyshows papilliferous proliferation with non-keratinised,hyperplastic epi<strong>the</strong>lium covering connectivetissue cores that may be patchily inflamed. Similar lesionshave been described in <strong>the</strong> absence <strong>of</strong> cutaneouslesions, usually in <strong>the</strong> midline <strong>of</strong> <strong>the</strong> palate.3.4.5 Smoker’s KeratosisKeratosis may be associated with both tobacco-smoking<strong>and</strong> -chewing. In smoker’s keratosis <strong>the</strong> lesions appear tobe a result <strong>of</strong> both <strong>the</strong>rmal <strong>and</strong> chemical irritation. Theyrarely show significant dysplasia <strong>and</strong> appear to have alow pre-malignant potential. The affected mucosa mayshow diffuse whitening or more focal lesions <strong>and</strong> occasionally<strong>the</strong>y are pigmented <strong>and</strong> have a slate-blue colour.The epi<strong>the</strong>lium can be hyperplastic or atrophic <strong>and</strong>is not dysplastic [136]. There is very variable parakeratosisor hyperorthokeratosis. Some cases show focalparakeratotic spikes (“chevrons”). Although suchchevrons were thought to be most characteristic <strong>of</strong>smokeless tobacco-induced keratosis, only a minority<strong>of</strong> such lesions show this feature [40]. Pigmentary incontinenceis common <strong>and</strong> may be florid in darkly pigmentedindividuals. Inflammatory infiltration is usuallyminimal.3.4.6 Stomatitis NicotinaStomatitis nicotina is usually seen in <strong>the</strong> palates <strong>of</strong> pipeor cigar smokers <strong>and</strong> <strong>the</strong> overwhelming majority <strong>of</strong> patientsare men [158]. Minor degrees <strong>of</strong> <strong>the</strong> condition maybe seen in heavy cigarette smokers. Similar clinical appearanceshave been reported in patients who regularlydrink excessively hot liquids. It is usually painless <strong>and</strong>asymptomatic. The initial lesion appears to be increased


Oral Cavity Chapter 3 85keratosis <strong>of</strong> <strong>the</strong> palate exposed to <strong>the</strong> smoke. This leadsto obstruction <strong>of</strong> <strong>the</strong> ducts <strong>of</strong> <strong>the</strong> underlying minor salivarygl<strong>and</strong>s, which <strong>the</strong>n become inflamed. The classicalclinical appearance, <strong>the</strong>refore, is whitening <strong>of</strong> <strong>the</strong> palatalmucosa, which may show tessellated plaque formation.The involved minor gl<strong>and</strong>s become swollen <strong>and</strong>have red, umbilicated centres.Microscopy shows variable hyperkeratosis, acanthosis<strong>and</strong> duct dilatation. There is usually no evidence <strong>of</strong>epi<strong>the</strong>lial dysplasia. There is variable submucosal chronicinflammation <strong>and</strong> <strong>the</strong>re may be evidence <strong>of</strong> pigmentaryincontinence. Keratinisation can extend down <strong>the</strong>salivary ducts <strong>and</strong> <strong>the</strong>re is interstitial inflammation <strong>of</strong><strong>the</strong> underlying minor mucous gl<strong>and</strong>s.The condition will gradually resolve if <strong>the</strong> habit isdiscontinued <strong>and</strong> <strong>the</strong>re appears to be minimal risk <strong>of</strong>malignant transformation. However, affected individualshave an increased risk <strong>of</strong> developing squamouscell carcinoma in o<strong>the</strong>r parts <strong>of</strong> <strong>the</strong> mouth, particularly<strong>the</strong> floor <strong>of</strong> <strong>the</strong> mouth <strong>and</strong> adjacent ventral lingualmucosa, <strong>and</strong> <strong>the</strong> retromolar trigone. Conversely, palatalkeratosis due to “reverse smoking” where <strong>the</strong> lightedend <strong>of</strong> a cigarette or cigar is held in <strong>the</strong> mouth isassociated with <strong>the</strong> development <strong>of</strong> carcinomas <strong>of</strong> <strong>the</strong>hard or s<strong>of</strong>t palates in a very high number <strong>of</strong> patientspractising <strong>the</strong> habit [148].3.4.7 Hairy TongueHairy tongue is due to hyperplasia <strong>and</strong> elongation <strong>of</strong><strong>the</strong> filiform papillae, which form hair-like overgrowthson <strong>the</strong> dorsum. The filaments can be several millimetreslong. The colour varies from pale brown to intenseblack. The discoloration is due to proliferation <strong>of</strong>chromogenic bacteria <strong>and</strong> fungi. Hairy tongue is usuallyseen in older individuals, <strong>and</strong> smoking, antisepticmouthwashes, antibiotics <strong>and</strong> a diet lacking abrasivefoodstuffs are <strong>the</strong> most common predisposing factors.The dorsum <strong>of</strong> <strong>the</strong> tongue may also become blackenedwithout elongation <strong>of</strong> <strong>the</strong> filiform papillae byantibiotic mouthwashes such as tetracycline <strong>and</strong> ironcompounds. Hairy tongue is rarely biopsied. Microscopically,it is characterised by irregular, hyperplasticfiliform papillae showing hyperorthokeratosis or hyperparakeratosiswith numerous bacterial conglomerates<strong>and</strong> filamentous organisms in <strong>the</strong> surface layers<strong>and</strong> more deeply between fronds <strong>of</strong> epi<strong>the</strong>lium. It hasbeen shown by immunocytochemical analysis <strong>of</strong> keratinexpression that in black hairy tongue <strong>the</strong>re is defectivedesquamation <strong>of</strong> cells in <strong>the</strong> central column <strong>of</strong><strong>the</strong> filiform papillae. This results in <strong>the</strong> typical highlyelongated, cornified spines that are <strong>the</strong> characteristicfeature <strong>of</strong> <strong>the</strong> condition [110].3.4.8 Hairy LeukoplakiaPatients infected with HIV, particularly homosexualmales, may develop characteristic intraoral white lesions[149]. The lateral margins <strong>and</strong> underlying ventrum <strong>of</strong><strong>the</strong> tongue are <strong>the</strong> most common sites. The lesion wascalled hairy leukoplakia, but typically it forms painless,vertical, white corrugations that may or may not havea rough or “hairy” surface. Some lesions are flat whiteplaques. O<strong>the</strong>r sites may also be involved, especially <strong>the</strong>post-commissural buccal mucosa.Microscopy shows acanthosis <strong>and</strong> parakeratosis,usually with verruciform, hair-like surface projections[168]. Invasion <strong>of</strong> <strong>the</strong> surface epi<strong>the</strong>lium by c<strong>and</strong>idal hyphaeis common. Immediately below <strong>the</strong> parakeratoticlayer <strong>the</strong>re is a zone <strong>of</strong> vacuolated <strong>and</strong> enlarged epi<strong>the</strong>lialcells with intense basophilic, pyknotic nuclei <strong>and</strong> perinuclearclearing (koilocytes). Epstein-Barr capsid viralantigen <strong>and</strong> viral particles can be demonstrated in <strong>the</strong>koilocytic nuclei [172]. There is usually little or no inflammatoryinfiltration <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium or underlyingcorium. Similar lesions have occasionally been reportedin patients receiving immunosuppressant drugs followingorgan transplantation. The early cases <strong>of</strong> hairy leukoplakiaassociated with HIV infection showed a veryhigh rate <strong>of</strong> progression to full-blown AIDS. Lesions canresolve spontaneously <strong>and</strong> usually respond well to antiviralor anti-retroviral drug treatment; <strong>the</strong>y appear tohave no premalignant potential.3.4.9 Geographic TongueGeographic tongue is a relatively common idiopathiccondition typically characterised by migrating areas <strong>of</strong>depapillation on <strong>the</strong> dorsum <strong>of</strong> <strong>the</strong> tongue [6]. In manycases it is associated with fissuring. There is loss <strong>of</strong> filiformpapillae <strong>of</strong>ten surrounded by a slightly raisedyellowish-white <strong>and</strong> crenellated margin. These areas<strong>of</strong> depapillation tend to heal centrally <strong>and</strong> spread centrifugally.Occasionally, <strong>the</strong> ventrum is involved <strong>and</strong> inthat site lesions consist <strong>of</strong> an area <strong>of</strong> ery<strong>the</strong>ma completelyor partially surrounded by a circinate whitish halo.Identical lesions can occasionally be seen elsewhere in<strong>the</strong> mouth <strong>and</strong> have been called “ectopic geographicaltongue”, although geographical stomatitis or benignmigratory stomatitis would be more appropriate terms[81]. The majority <strong>of</strong> cases <strong>of</strong> geographical tongue arepainless, but some patients complain bitterly <strong>of</strong> soreness<strong>and</strong> discomfort, which may or may not be associatedwith specific foods.Geographical tongue is usually obvious clinically<strong>and</strong> is rarely biopsied. However, it has very typical microscopicalfeatures [114]. There is loss <strong>of</strong> filiform papillae<strong>and</strong> typically only a mild chronic inflammatory


86 J.W. Eveson3shredded, <strong>of</strong>ten patchily ery<strong>the</strong>matosus white plaqueslimited to areas accessible to chewing. In typical frictionalkeratosis microscopy usually shows a thick orthokeratinisedlayer with a prominent granular cell layer.There is no significant dysplasia <strong>and</strong> inflammation <strong>of</strong><strong>the</strong> underlying corium may be minimal in <strong>the</strong> absence<strong>of</strong> ulceration. In keratosis due to habitual chewing <strong>the</strong>reis <strong>of</strong>ten acanthosis <strong>and</strong> <strong>the</strong> surface is usually irregular<strong>and</strong> parakeratinised. It frequently shows a covering <strong>of</strong>adherent basophilic cocci or more dense bacterial conglomerates.Fig. 3.11. Geographical tongue (ery<strong>the</strong>ma migrans) showingpolymorphonuclear leukocyte microabscesses in <strong>the</strong> epi<strong>the</strong>lium3.5 Pigmentationsreaction in <strong>the</strong> underlying corium. The striking featureis <strong>the</strong> presence <strong>of</strong> polymorphonuclear leukocytic microabscessesin <strong>the</strong> upper stratum spinosum (Fig. 3.11).These spongiform pustules are not pathognomonic <strong>of</strong>geographical tongue <strong>and</strong> can be seen in oral psoriasis,acute <strong>and</strong> chronic c<strong>and</strong>idosis, Reiter syndrome <strong>and</strong> plasmacell gingivostomatosis. Some describe elongation <strong>of</strong><strong>the</strong> rete ridges, but this is by no means a consistent observation.However, occasionally <strong>the</strong>re may be psoriasiformhyperplasia in geographical tongue <strong>and</strong> it may bedifficult or impossible to distinguish from psoriasis. Indeed,geographical tongue <strong>and</strong> migratory stomatitis are4–5 times more common in patients with psoriasis <strong>and</strong>some believe that geographical tongue is <strong>the</strong> oral homology<strong>of</strong> psoriasis [179, 194]. The presence <strong>of</strong> spongiformpustules in oral biopsies should always prompt<strong>the</strong> search for c<strong>and</strong>idal hyphae with a PAS or Grocottstain. These are not usually seen in geographical tongue<strong>and</strong> <strong>the</strong>ir presence would make a diagnosis <strong>of</strong> c<strong>and</strong>idosismuch more likely. Chronic hyperplastic c<strong>and</strong>idosis canalso show psoriasiform hyperplasia, but typically <strong>the</strong>reis much more florid inflammatory infiltration <strong>of</strong> <strong>the</strong> corium<strong>and</strong> irregular surface parakeratosis.3.4.10 Frictional KeratosisFrictional keratosis <strong>of</strong> <strong>the</strong> oral mucosa is common <strong>and</strong> isusually a response to low-grade irritation by such causesas sharp edges <strong>of</strong> teeth or restorations, dental pros<strong>the</strong>ses,abrasive foods, vigorous tooth brushing <strong>and</strong> playingwind instruments.Clinically, <strong>the</strong> lesions tend to form diffuse keratoticplaques. In <strong>the</strong> early stages <strong>the</strong>se are pale <strong>and</strong> translucent<strong>and</strong> merge imperceptibly into <strong>the</strong> surrounding normalmucosa. Later, <strong>the</strong>y become more dense <strong>and</strong> white<strong>and</strong> may have an irregular, shaggy surface. The habit <strong>of</strong>cheek, lip <strong>and</strong> tongue chewing is a usually characteristicvariant <strong>of</strong> frictional keratosis. It results in roughened or3.5.1 Amalgam TattooAmalgam tattoo is <strong>the</strong> most common localised form <strong>of</strong>oral mucosal pigmentation [23]. Lesions usually formpainless, bluish-black macules, which may be well definedor diffuse, <strong>and</strong> <strong>the</strong> most common sites are <strong>the</strong> gingiva,alveolar mucosa <strong>and</strong> floor <strong>of</strong> <strong>the</strong> mouth. They arecaused by ingress <strong>of</strong> dental amalgam through a mucosalbreach during a restorative procedure or tooth extraction<strong>and</strong> can also follow an apicectomy with retrograderoot filling.Microscopy shows dark, refractile particles <strong>of</strong> amalgamin <strong>the</strong> corium [44]. These may be coarse, but usuallyform fine, black or brownish granules. These aredeposited along collagen <strong>and</strong> elastic fibres, <strong>and</strong> aroundsmall blood vessels, nerves <strong>and</strong> muscles. About half <strong>of</strong>cases show a fibrous <strong>and</strong> chronic inflammatory reaction,with or without multinucleated foreign body giantcells. Occasionally, <strong>the</strong>re is a granulomatous reaction.3.5.2 Localised Melanotic Pigmentation3.5.2.1 Oral Melanotic MaculesThese benign, ephelis-like pigmented macules are <strong>the</strong>most common melanocytic lesions <strong>of</strong> <strong>the</strong> oral mucosa.They are brownish-blue or black <strong>and</strong> may be single ormultiple. They are typically well defined <strong>and</strong> rarelyexceed 6 mm in diameter. They develop during earlyto middle adult life with a mean age at presentation <strong>of</strong>41 years [22]. There is a female predilection <strong>of</strong> 2:1. Theyare most frequent in <strong>the</strong> anterior part <strong>of</strong> <strong>the</strong> mouth affecting<strong>the</strong> gingiva, buccal mucosa <strong>and</strong>, most commonly,<strong>the</strong> labial mucosa. Those involving <strong>the</strong> vermilion border(labial melanotic macules) <strong>of</strong>ten darken in strong sunlight<strong>and</strong> may cause cosmetic problems. Oral melanoticmacules have been reported following radio<strong>the</strong>rapy [11],


Oral Cavity Chapter 3 87Fig. 3.12. Melanotic macule showing increased melanotic pigmentation<strong>of</strong> <strong>the</strong> basal keratinocytes <strong>and</strong> melanophages in <strong>the</strong> superficialcorium<strong>and</strong> in HIV infections, probably related to <strong>the</strong> administration<strong>of</strong> retroviral drugs [55].Microscopy shows increased melanotic pigmentationin <strong>the</strong> basal <strong>and</strong> occasionally <strong>the</strong> immediately suprabasalkeratinocytes. There is <strong>of</strong>ten pigmentary incontinence<strong>and</strong> melanophages in <strong>the</strong> superficial corium(Fig. 3.12).3.5.2.2 MelanoacanthomaMelanoacanthoma (melanoacanthosis) is a rare, probablyreactive, proliferation <strong>of</strong> both keratinocytes <strong>and</strong>melanocytes [61, 64]. It is seen most commonly in adultblack females, typically involving <strong>the</strong> labial or buccalmucosa. Lesions may be single or multiple [49] <strong>and</strong> areusually macular; less frequently, <strong>the</strong>y are slightly raisedor papilliferous. Trauma is thought to be <strong>the</strong> most likelycause <strong>and</strong> lesions can regress spontaneously or followingincisional biopsy.Microscopy shows acanthosis <strong>and</strong> frequently spongiosisthat can be florid. There are strongly Fontana silverstain- <strong>and</strong> HMB45-positive, dendritic melanocytesextending throughout <strong>the</strong> full thickness <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium(Fig. 3.13). As a consequence <strong>of</strong> a partial or completeblock in pigment transfer <strong>the</strong> keratinocytes in melanoacanthomacontain little or no melanin in spite <strong>of</strong> <strong>the</strong>abundance <strong>of</strong> melanocytes.3.5.2.3 Pigmented NaeviOral melanocytic naevi are much rarer than cutaneouslesions with a prevalence <strong>of</strong> 0.1% <strong>of</strong> <strong>the</strong> general population[75]. The most common sites are <strong>the</strong> hard palate,buccal mucosa <strong>and</strong> labial mucosa. Women are affectedtwice as frequently as men <strong>and</strong> most cases are seen in <strong>the</strong>third <strong>and</strong> fourth decades. They are usually single <strong>and</strong>Fig. 3.13. Melanoacanthoma showing dendritic melanocytes extendingthroughout <strong>the</strong> epi<strong>the</strong>lial thickness. Inset shows melanocytesstained with Masson-Fontanaform brown, bluish or black macules or sessile papules.Most are less than 6 mm in diameter [24]. Intramucosal(intradermal) naevi account for over half <strong>of</strong> <strong>the</strong> cases<strong>and</strong> junctional or compound naevi are uncommon. Bluenaevi form 25–35% <strong>of</strong> cases <strong>and</strong> usually present in <strong>the</strong>palate. The large majority are <strong>of</strong> <strong>the</strong> common ra<strong>the</strong>rthan <strong>the</strong> cellular type.3.5.3 Premalignant Oral Melanoses<strong>and</strong> Oral MelanomaICD-O:8720/3Oral mucosal melanomas are rare <strong>and</strong> account for about0.5% <strong>of</strong> oral malignancies [35, 75]. However, <strong>the</strong>y appearto be more common in Japan, India <strong>and</strong> Africa. Mostarise in adults with a peak incidence between 40 <strong>and</strong>60 years. Large series show a male predominance. About80% <strong>of</strong> cases involve <strong>the</strong> palate <strong>and</strong> maxillary alveolus<strong>and</strong> gingivae. About a third <strong>of</strong> cases are preceded by


88 J.W. Eveson3long-st<strong>and</strong>ing areas <strong>of</strong> oral hyperpigmentation, but <strong>the</strong>yrarely arise from pre-existing benign melanocytic naevi.The majority <strong>of</strong> cases are painless in <strong>the</strong> early stages <strong>and</strong>form irregular, black or brownish flat, raised or nodularareas that are frequently multicentric. Rarely, <strong>the</strong>y areamelanotic <strong>and</strong> may be reddish in colour. Nodular areasare usually a feature <strong>of</strong> more advanced tumours <strong>and</strong>may be ulcerated <strong>and</strong> associated with pain <strong>and</strong> bleeding.Invasion <strong>of</strong> <strong>the</strong> underlying bone is common <strong>and</strong> teethinvolved may loosen or exfoliate. In most cases <strong>the</strong>re isinvolvement <strong>of</strong> <strong>the</strong> cervical lymph nodes at presentation<strong>and</strong> half <strong>of</strong> patients have distant metastases.Purely nodular melanomas are relatively rare <strong>and</strong>most tumours have a radial growth element similar tothat seen in cutaneous acral lentiginous melanoma toge<strong>the</strong>rwith evidence <strong>of</strong> upward migration. Oral melanomashave been divided into:1. In situ oral mucosal melanomas;2. Invasive oral mucosa melanomas;3. Mixed in situ <strong>and</strong> invasive oral mucosal melanomas.Fig. 3.14. In situ melanoma showing atypical melanocytes widelydispersed throughout <strong>the</strong> epi<strong>the</strong>liumAbout 15% <strong>of</strong> oral mucosal melanomas are in situ <strong>and</strong>30% are invasive [9]. Fifty-five percent <strong>of</strong> melanomashave a combined pattern. Borderline lesions have beentermed atypical melanocytic proliferations [75].Microscopically, in situ melanomas show an increasein atypical melanocytes. Although <strong>the</strong>se atypical melanocyteshave angular <strong>and</strong> hyperchromatic nuclei, mitosestend to be sparse. The melanocytes may form aggregatesor be irregularly distributed in a junctional location.The characteristic nested or <strong>the</strong>qual pattern commonlyseen in cutaneous melanomas is less frequentlyobserved in mucosal lesions. Sometimes <strong>the</strong> melanocytesare dispersed throughout <strong>the</strong> epi<strong>the</strong>lium <strong>and</strong> thismay be combined with a junctional pattern (Fig. 3.14).Sequential biopsies have shown increases in <strong>the</strong> density<strong>of</strong> <strong>the</strong> junctional atypical melanocytes over time. Atypicalmelanocytes can extend down <strong>the</strong> excretory ducts <strong>of</strong><strong>the</strong> underlying minor salivary gl<strong>and</strong>s. However, <strong>the</strong>re isusually no inflammatory response to in situ lesions.The melanocytes present in invasive melanomasshow a variety <strong>of</strong> cell types including epi<strong>the</strong>lioid, spindle<strong>and</strong> plasmacytoid. They typically have large, vesicularnuclei with prominent nucleoli. Mitoses may be present,but usually not in large numbers. They are usuallyaggregated into sheets or alveolar groups <strong>and</strong> less commonlyneurotropic or desmoplastic configurations areseen. About 10% <strong>of</strong> cases are amelanotic. Over 95% <strong>of</strong> lesionsare anti S-100 antigen-positive [10] <strong>and</strong> more specificmarkers include HMB45, Melan-A <strong>and</strong> antityrosinase[144].Atypical melanocytic proliferation or hyperplasia is<strong>the</strong> term used for lesions with equivocal histopathologicalfeatures, but <strong>the</strong> criteria for inclusion in this categoryare ra<strong>the</strong>r ill-defined [176]. These include oral mucosallesions with melanocytes containing angular or hyperchromaticnuclei with very infrequent mitoses. Melanocyticatypia can vary from mild to severe [75].Oral melanomas are much more aggressive than <strong>the</strong>ircutaneous counterparts. The prognosis <strong>of</strong> oral melanomasis poor with a 5-year survival rate <strong>of</strong> less than 20%[9], <strong>and</strong> even Stage I tumours have a 5-year survival rate<strong>of</strong> less than 50%. The conventional depth <strong>of</strong> invasionindicators such as Breslow thickness <strong>and</strong> Clark’s levelstend to be <strong>of</strong> little value in mucosal melanomas, as manypresent at an advanced stage <strong>and</strong> most are deeper than4 mm [145]. Histological features associated with a poorprognosis include evidence <strong>of</strong> vascular invasion, cellularpleomorphism, necrosis <strong>and</strong> amelanotic tumours [14,75, 123, 145].3.5.4 Addison DiseaseAddison disease is rare with an estimated annual incidence<strong>of</strong> 0.8 cases per 100,000 <strong>of</strong> <strong>the</strong> population inWestern societies. It is due to bilateral destruction <strong>of</strong> <strong>the</strong>adrenal cortex. Formerly, <strong>the</strong> most common cause wastuberculosis. Most cases now are due to organ-specificauto-immune destruction <strong>and</strong> opportunistic infectionssuch as histoplasmosis in patients with AIDS. Due tothis, it is likely that <strong>the</strong> number <strong>of</strong> patients with Addisondisease will increase significantly. Addison diseasemay be associated with autoimmune polygl<strong>and</strong>ular deficiencytype I (Addison disease, chronic mucocutaneousc<strong>and</strong>idosis, hypoparathyroidism) <strong>and</strong> autoimmunepolygl<strong>and</strong>ular deficiency type II (Addison disease, primaryhypothyroidism, primary hypogonadism, insulindependentdiabetes, pernicious anaemia, vitiligo) [130].Clinically, <strong>the</strong>re is usually slowly progressive weakness,lassitude <strong>and</strong> weight loss. Gastro-intestinal symptomscan include diarrhoea or constipation <strong>and</strong> anorexia,nausea <strong>and</strong> vomiting. Postural hypotension is a commonsymptom. An early sign is pigmentation <strong>of</strong> <strong>the</strong> skin<strong>and</strong> oral mucosa secondary to increased adrenocortico-


Oral Cavity Chapter 3 89tropic hormone (ACTH) secretion. Parts <strong>of</strong> <strong>the</strong> skin exposedto <strong>the</strong> sun <strong>and</strong> areas subjected to trauma or frictionbecome bronzed [85]. There is also increased pigmentationin skin folds <strong>and</strong> scars. About 10% <strong>of</strong> patientsalso show areas <strong>of</strong> vitiligo.Oral pigmentation is variable <strong>and</strong> where presentranges from light brown to densely black [95]. The gingiva,lateral margins <strong>of</strong> <strong>the</strong> tongue, buccal mucosa <strong>and</strong>lips are <strong>the</strong> sites <strong>of</strong> predilection. Microscopy <strong>of</strong> <strong>the</strong> areasshows increased melanin predominantly in <strong>the</strong> basalkeratinocytes.3.5.5 Peutz Jeghers SyndromePeutz Jeghers syndrome (periorifacial lentiginosis)comprises melanotic spots <strong>of</strong> <strong>the</strong> face, mouth <strong>and</strong> lesscommonly <strong>the</strong> h<strong>and</strong>s <strong>and</strong> feet, toge<strong>the</strong>r with intestinalpolyposis. It is inherited as an autosomal dominant traitwith nearly complete penetrance [93], but new mutationsoccur in 40% <strong>of</strong> cases.There are multiple ephelides on <strong>the</strong> face <strong>and</strong> melanoticmacules in <strong>the</strong> mouth involving <strong>the</strong> lower labial<strong>and</strong> buccal mucosa in particular. Lesions are <strong>of</strong>tenpresent at birth. The facial pigmentation is around <strong>the</strong>mouth, nose <strong>and</strong> eyes <strong>and</strong> tends to progressively fadeafter puberty. The mucosal pigmentation persists intoadult life.There are hamartomatous polyps throughout <strong>the</strong> intestinaltract, but typically <strong>the</strong>se are most numerous in<strong>the</strong> small intestine. They can give rise to abdominal pain<strong>and</strong> bleeding <strong>and</strong> intussusception is a rare complication.The polyps have a low malignant potential, with thosein <strong>the</strong> colon having <strong>the</strong> highest risk. Patients also are atincreased risk <strong>of</strong> malignancy at o<strong>the</strong>r sites including <strong>the</strong>uterus, ovary, pancreas <strong>and</strong> breast.3.5.6 Racial PigmentationRacial pigmentation is <strong>the</strong> most common cause <strong>of</strong> intraoralpigmentation. It is seen predominantly in Blacks,Asians <strong>and</strong> people <strong>of</strong> Mediterranean origin, but about5% <strong>of</strong> Caucasians also have significant intraoral pigmentation.The degree <strong>and</strong> extent <strong>of</strong> racial pigmentationis very variable <strong>and</strong> does not necessarily correlate with<strong>the</strong> depth <strong>of</strong> skin pigmentation. It can vary from lightbrown to almost black <strong>and</strong> <strong>the</strong> most commonly involvedsites are <strong>the</strong> gingiva, palate <strong>and</strong> buccal mucosa. Sometimeswhen <strong>the</strong> tongue is involved <strong>the</strong> only areas affectedare <strong>the</strong> fungiform papillae, producing an appearance<strong>of</strong> spotty pigmentation.Microscopy shows increased melanocytic pigmentation<strong>of</strong> <strong>the</strong> basal <strong>and</strong>, to a much lesser degree, <strong>the</strong> immediatelysuprabasal keratinocytes. The denser pigmentationis due to increased syn<strong>the</strong>sis <strong>of</strong> melanin by melanocytes,which are o<strong>the</strong>rwise normal in number <strong>and</strong> distribution.3.5.7 Laugier Hunziker SyndromeLaugier Hunziker syndrome is an acquired, benignmacular hyperpigmentation <strong>of</strong> <strong>the</strong> lips <strong>and</strong> oral mucosa[118]. It typically starts in early to middle adult life <strong>and</strong>is more common in women than men. The pigmentationconsists <strong>of</strong> brownish, circular or linear macules,that may be sharply circumscribed or more diffuse innature, <strong>and</strong> lesions may coalesce. The lesions are seenmost commonly on <strong>the</strong> buccal mucosa, lips <strong>and</strong> <strong>the</strong>hard <strong>and</strong> s<strong>of</strong>t palates. O<strong>the</strong>r less frequent sites include<strong>the</strong> tongue, gingiva <strong>and</strong> floor <strong>of</strong> <strong>the</strong> mouth. Occasionally,<strong>the</strong> pharynx <strong>and</strong> oesophagus are also involved[190]. About half <strong>of</strong> <strong>the</strong> cases also have nail involvementin <strong>the</strong> form <strong>of</strong> longitudinal pigmented b<strong>and</strong>s inone or more fingers or toes. There are no known systemicassociations.Microscopy shows increased melanotic pigmentation<strong>of</strong> basal keratinocytes <strong>and</strong> melanophages in <strong>the</strong> superficialcorium secondary to pigmentary incontinence. Ultrastructuralstudies show increased numbers <strong>of</strong> normal-appearingmelanosomes in keratinocytes in <strong>the</strong>lower epi<strong>the</strong>lial layers [118].3.5.8 Smoker’s MelanosisHeavy smokers can sometimes develop areas <strong>of</strong> oralhyperpigmentation. It is more common in womenthan men. Although any part <strong>of</strong> <strong>the</strong> mouth can beaffected <strong>the</strong> anterior gingivae are involved most frequently.The lesions vary in colour from light brownto bluish-black <strong>and</strong> <strong>the</strong> lesions may be focal or diffuse.Sometimes <strong>the</strong> overlying mucosa has a somewhatmilky-white appearance, particularly in <strong>the</strong> buccalmucosa. The condition can slowly resolve if smokingis stopped or reduced [72]. Pigmentation <strong>of</strong> <strong>the</strong> s<strong>of</strong>tpalate has been reported in a significant number <strong>of</strong>patients with suppurative lung disease <strong>and</strong> malignancy[117]. Nearly a quarter <strong>of</strong> patients with confirmedbronchogenic carcinoma show this feature. Most patientshave a long history <strong>of</strong> cigarette smoking <strong>and</strong> it ispossible that in many cases <strong>the</strong>se lesions were merelysmoker’s melanosis ra<strong>the</strong>r than being related directlyto <strong>the</strong> pulmonary lesions.Microscopy may show slightly increased melanoticpigmentation <strong>of</strong> <strong>the</strong> basal keratinocytes, but <strong>the</strong> moststriking feature is usually pigmentary incontinence <strong>and</strong>accumulation <strong>of</strong> melanophages in <strong>the</strong> superficial corium.


90 J.W. Eveson3.5.9 Drug-AssociatedOral Pigmentation3Gingival pigmentation due to heavy metals such asmercury, lead, bismuth, arsenic <strong>and</strong> o<strong>the</strong>rs was notrare in <strong>the</strong> past due to industrial exposure <strong>and</strong> in somecases <strong>the</strong>rapeutic administration, particularly for <strong>the</strong>treatment <strong>of</strong> syphilis. They caused blue, brown orblack lines close to <strong>the</strong> gingival margins due to <strong>the</strong> deposition<strong>of</strong> sulphides as a result <strong>of</strong> reactions with products<strong>of</strong> <strong>the</strong> dental plaque. A wide range <strong>of</strong> drugs cancause more generalised oral pigmentation includingantimalarials, phenothiazines <strong>and</strong> some contraceptivepills [98]. Drugs used in <strong>the</strong> treatment <strong>of</strong> HIV infectionsuch as zidovudine <strong>and</strong> some antifungals such asketoconazole have also been shown to cause oral pigmentation.3.6 Hyperplastic Lesions3.6.1 Fibrous HyperplasiasThe majority <strong>of</strong> fibrous <strong>and</strong> fibroblastic lesions within<strong>the</strong> mouth appear to be reactive ra<strong>the</strong>r than neoplastic.They are <strong>the</strong> most common tumour-like swelling <strong>of</strong> oralmucosa. Although <strong>the</strong>se lesions are considered to be aresponse to low-grade irritation, <strong>the</strong> source <strong>of</strong> such irritationmay not be immediately apparent. Fibroepi<strong>the</strong>lialpolyps tend to form smooth nodules or swellings thatmay be s<strong>of</strong>t or firm <strong>and</strong> are usually covered by normal,pink mucosa unless ulcerated. The polypoid swellingsmay be sessile or pedunculated.Fibrous overgrowths <strong>of</strong> <strong>the</strong> gingiva are a type <strong>of</strong> epulis(lit. – swelling <strong>of</strong> <strong>the</strong> gum). They can arise from <strong>the</strong>interdental papilla or gingival margin <strong>and</strong> tend to affect<strong>the</strong> anterior part <strong>of</strong> <strong>the</strong> mouth. They can grow to severalcentimetres in diameter. Fibrous epulides are frequentlyassociated with local irritation from dental calculus,sharp edges <strong>of</strong> restorations or carious teeth. A very characteristicform <strong>of</strong> hyperplasia is associated with <strong>the</strong> edges<strong>of</strong> loose dentures. Such denture-induced fibrous hyperplasiahas been termed denture granuloma <strong>and</strong> epulisfissuratum. The rocking backwards <strong>and</strong> forwards <strong>of</strong><strong>the</strong> denture causes extensive overgrowth <strong>of</strong> fibrous tissueon ei<strong>the</strong>r side <strong>of</strong> <strong>the</strong> edges, or flanges, <strong>of</strong> <strong>the</strong> denture.This <strong>of</strong>ten leads to <strong>the</strong> formation <strong>of</strong> a series <strong>of</strong> linearfolds <strong>of</strong> hyperplastic tissue <strong>and</strong> <strong>the</strong> base <strong>of</strong> <strong>the</strong> groovesso formed is <strong>of</strong>ten ulcerated by <strong>the</strong> denture’s edge. O<strong>the</strong>rcommon sites for fibrous overgrowths are along <strong>the</strong>occlusal line <strong>of</strong> <strong>the</strong> buccal mucosa <strong>and</strong> lateral border <strong>of</strong><strong>the</strong> tongue, <strong>and</strong> related to spaces where teeth have beenextracted.Microscopically most <strong>of</strong> <strong>the</strong>se nodules consist <strong>of</strong> interlacingbundles <strong>of</strong> sparsely cellular fibrous tissue. TheFig. 3.15. Fibrous epulis showing osseous metaplasiaoverlying epi<strong>the</strong>lium is <strong>of</strong>ten hyperplastic with irregularrete processes extending sometimes deeply into <strong>the</strong> underlyingfibrous tissue. There may be c<strong>and</strong>idal infestation<strong>of</strong> <strong>the</strong> superficial epi<strong>the</strong>lium. The degree <strong>of</strong> inflammatoryinfiltration is very variable, but tends to be mildunless <strong>the</strong>re has been ulceration.The microscopical appearances <strong>of</strong> fibrous epulidescan differ from fibrous overgrowths seen elsewhere in<strong>the</strong> mouth. They typically show much more evidence<strong>of</strong> cellular fibroblastic proliferation. These lesions mayconsist predominantly or focally <strong>of</strong> a vascular stromacontaining plump fibroblasts with large, vesicular nuclei<strong>and</strong> prominent nucleoli. There can be brisk mitoticactivity. Ulceration is common <strong>and</strong> <strong>the</strong> lesions are<strong>of</strong>ten heavily inflamed. Calcifications are common infibrous epulides <strong>and</strong> <strong>the</strong>re may be florid osseous metaplasiaor dystrophic calcification (Fig. 3.15). Sometimes<strong>the</strong> calcified masses are basophilic <strong>and</strong> <strong>the</strong>y canalso be laminated <strong>and</strong> resemble cementicles. Such lesionshave been termed “peripheral ossifying fibromas”,but <strong>the</strong>re is no evidence that <strong>the</strong>y are neoplasticor have any relationship with central ossifying fibromas(see Chap. 4). Mineralisation tends to be uncommonin extra-gingival oral fibrous overgrowths.Most fibrous overgrowths respond to conservativesurgical removal, but a minority <strong>of</strong> fibrous epulides canrecur, sometimes repeatedly.3.6.2 Papillary HyperplasiaPapillary hyperplasia is typically seen in <strong>the</strong> hard palate.In many cases it is related to dentures as part <strong>of</strong> <strong>the</strong>clinical spectrum <strong>of</strong> denture-induced stomatitis [174].Although C<strong>and</strong>ida albicans is frequently invoked as <strong>the</strong>causal agent, in a significant number <strong>of</strong> cases <strong>the</strong>re is noevidence <strong>of</strong> fungal infection. The large majority <strong>of</strong> casesinvolve <strong>the</strong> hard palate, particularly when this is higharched,but similar lesions are occasionally seen on <strong>the</strong>dorsum <strong>of</strong> <strong>the</strong> tongue. The lesions form painless, nodu-


Oral Cavity Chapter 3 91Fig. 3.16. Papillary hyperplasia <strong>of</strong> <strong>the</strong> palate showing long reteprocesses extending into <strong>the</strong> connective tissue coreslar or papilliferous proliferations. Florid cases have beenreported in immunocompromised patients [151].Microscopy shows nodular, papilliferous hyperplasia<strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium <strong>and</strong> underlying fibrous connectivetissue (Fig. 3.16). The surface usually shows parakeratosisor less commonly orthokeratosis. There may be evidence<strong>of</strong> c<strong>and</strong>idal infestation such as spongiform pustulesor obvious hyphae. The underlying hyperplasticrete ridges <strong>of</strong>ten extend into <strong>the</strong> cores <strong>of</strong> <strong>the</strong> papillae, resultingin a striking pseudoepi<strong>the</strong>liomatous appearance.The corium <strong>of</strong>ten contains a dense chronic inflammatorycell infiltrate. The condition <strong>the</strong>n needs to be distinguishedfrom oral papillary plasmacytosis [66].3.6.3 GeneralisedGingival Fibrous HyperplasiaGeneralised fibrous hyperplasia/hypertrophy <strong>of</strong> <strong>the</strong>gingiva can be familial or drug-induced.Hereditary gingival fibromatosis is a rare conditionthat is usually inherited as an autosomal dominant trait[20]. It can affect all <strong>of</strong> <strong>the</strong> gingiva, <strong>of</strong>ten in a symmetricalmanner. It may be associated with hypertrichosis,coarsening <strong>of</strong> <strong>the</strong> facial features <strong>and</strong> neurological problemssuch as epilepsy <strong>and</strong> mental retardation. The conditionusually first affects adolescents, but occasionallyit can involve <strong>the</strong> deciduous dentition. The enlargementis usually most conspicuous in <strong>the</strong> interdental areas <strong>and</strong>affects <strong>the</strong> palatal <strong>and</strong> lingual gingiva as well as <strong>the</strong> labial<strong>and</strong> buccal aspects. The overgrowths may be so floridthat <strong>the</strong> teeth involved are almost completely buried(pseudo-anodontia). The overgrowths are rounded,smooth, firm <strong>and</strong> pale-coloured. Treatment is by surgicalremoval <strong>of</strong> <strong>the</strong> redundant fibrous tissue (gingivectomy),but <strong>the</strong> condition <strong>of</strong>ten recurs.Drug-associated gingival hypertrophy is seen in abouthalf <strong>of</strong> patients using <strong>the</strong> anti-epileptic drug phenytoinfor long-term treatment [1]. O<strong>the</strong>r drugs producing asimilar reaction include cyclosporin (~30% <strong>of</strong> patients)<strong>and</strong> calcium channel blockers such as nifedipine, amlodipine<strong>and</strong> verapamil (~10% patients). The enlargementsmay be generalised or more localised. The anteriorinterdental papillae tend to be <strong>the</strong> most severely affectedareas, particularly on <strong>the</strong> labial aspect. The gingivatend to enlarge laterally <strong>and</strong> this growth pattern mayresult in <strong>the</strong> formation <strong>of</strong> vertical clefts between adjacentovergrowths. The normal gingival stippling may beenhanced, producing an orange-peel appearance. Thecondition is exacerbated by poor oral hygiene <strong>and</strong> meticulousattention to tooth cleaning may help prevent<strong>the</strong> development <strong>of</strong> <strong>the</strong> condition.Microscopy <strong>of</strong> generalised gingival fibrous hyperplasiatends to be <strong>the</strong> same irrespective <strong>of</strong> <strong>the</strong> cause. Thereis fibrous hypertrophy <strong>of</strong> <strong>the</strong> affected gingiva, <strong>of</strong>ten containingmy<strong>of</strong>ibroblastic cells. An increase in <strong>the</strong> amount<strong>of</strong> myxoid ground substance material is also common.In areas distant from <strong>the</strong> gingival sulcus <strong>the</strong>re is usuallyno significant inflammatory component. There is <strong>of</strong>tenelongation <strong>and</strong> fusing <strong>of</strong> <strong>the</strong> rete process <strong>of</strong> <strong>the</strong> overlyingepi<strong>the</strong>lium. It is unusual for <strong>the</strong>se lesions to becomeulcerated3.6.4 Crohn’s DiseaseCrohn’s disease is a multisystem disorder characterisedmicroscopically by non-caseating, epi<strong>the</strong>lioid granulomas.Despite extensive investigations <strong>the</strong> cause remainsunknown. Recently, genetic predisposition has been extensivelyinvestigated <strong>and</strong> among o<strong>the</strong>rs, a susceptibilitygene has been identified on chromosome 16q [18].Smoking also appears to play a critical role in some patients[60]. Although an infective aetiology, particularlymycobacterial, has been long suspected, critical pro<strong>of</strong>is lacking [173]. The most likely c<strong>and</strong>idate organism is<strong>the</strong> Mycobacterium avium subspecies paratuberculosis[68]. Granulomatous vasculitis, possibly initiated by <strong>the</strong>measles virus, has been proposed as a significant factorin <strong>the</strong> pathogenesis [178]. However, blood vessel involvementmay be a secondary phenomenon ra<strong>the</strong>r thana primary event [120]. Patients are usually children <strong>and</strong>young adults <strong>and</strong> <strong>the</strong>re is a male preponderance. Associationsbetween alterations in <strong>the</strong> intestinal micr<strong>of</strong>loraare also suspected <strong>and</strong> a statistically significant associationbetween Crohn’s disease <strong>and</strong> previous antibioticuse has been proposed [29]. The condition may be madeworse by cigarette smoking [80].Oral lesions are relatively common in patients withCrohn’s disease <strong>of</strong> <strong>the</strong> lower gastro-intestinal tract [57,185], but may be <strong>the</strong> presenting symptoms. They includeswelling <strong>of</strong> <strong>the</strong> lips <strong>and</strong> cheeks (Fig. 3.17), recurrent aphthae,painful, indolent linear ulcers in <strong>the</strong> vestibular sulci,cobblestone thickening <strong>of</strong> <strong>the</strong> buccal mucosa, mucosaltags <strong>and</strong> hyperplastic, granular gingivitis [138]. Thepalate, tongue <strong>and</strong> pharynx, including <strong>the</strong> palatine ton-


92 J.W. Eveson3Fig. 3.17. Crohn’s disease showing unilateral lip swelling with fissuring<strong>and</strong> peri-oral dermatitisFig. 3.18. Crohn’s disease showing a small, irregular epi<strong>the</strong>lioidgranuloma <strong>and</strong> patchy chronic inflammationsil, are only rarely involved [19]. Extraorally, <strong>the</strong>re maybe angular stomatitis <strong>and</strong> vertical fissuring <strong>of</strong> <strong>the</strong> lips,<strong>and</strong> perioral ery<strong>the</strong>ma <strong>and</strong> scaling.Oral lesions may precede, or accompany, bowel symptoms,but in a significant number <strong>of</strong> cases intestinal diseaseis subclinical. In patients with active bowel disease<strong>the</strong>re may be atrophic glossitis secondary to malabsorption<strong>of</strong> <strong>the</strong> haematinics iron, vitamin B 12 or folate.Microscopy <strong>of</strong> oral lesions typically shows oedema<strong>of</strong> <strong>the</strong> superficial corium with lymphangiectasia<strong>and</strong> diffuse <strong>and</strong> focal aggregates <strong>of</strong> small lymphocytes.Non-caseating epi<strong>the</strong>lioid granulomas with orwithout multinucleated giant cells are present in about90% <strong>of</strong> cases [138]. However, granulomas may be small<strong>and</strong> poorly formed <strong>and</strong> may only be present in <strong>the</strong> underlyingmuscle so that <strong>the</strong>y can be easily missed, especiallyif <strong>the</strong> biopsy is superficial (Fig. 3.18). Granulomascan also sometimes be seen in <strong>the</strong> minor salivarygl<strong>and</strong>s. Aggregates <strong>of</strong> mononuclear cells or granulomasmay be seen bulging into or within <strong>the</strong> lumina<strong>of</strong> lymphatics. This feature has been termed endovasalgranulomatous lymphangiitis [128]. Dilated lymphatics,with or without associated granulomas, are characteristic<strong>of</strong> Crohn’s disease elsewhere in <strong>the</strong> alimentarytract [120].3.6.5 Or<strong>of</strong>acial GranulomatosisAs many as 80–90% <strong>of</strong> patients with or<strong>of</strong>acial lesionsthat resemble those <strong>of</strong> Crohn’s disease, both clinically<strong>and</strong> microscopically, have no gastro-intestinal signs orsymptoms <strong>and</strong> do not develop gut disease [159]. Theterm or<strong>of</strong>acial granulomatosis (OFG) has been introducedto describe this group <strong>of</strong> patients [184]. OFG,<strong>the</strong>refore, is a diagnosis based on <strong>the</strong> exclusion <strong>of</strong> o<strong>the</strong>rcauses <strong>of</strong> granulomatous inflammation, particularlysarcoidosis, tuberculosis <strong>and</strong> o<strong>the</strong>r mycobacterial infections,<strong>and</strong> Crohn’s disease itself. Up to 60% <strong>of</strong> patientswith OFG are atopic [86] <strong>and</strong> some patients appear toshow an idiosyncratic intolerance to a variety <strong>of</strong> foodsor additives, including cinnamonaldehyde, carvone,carnosine, sun yellow, benzoates <strong>and</strong> monosodium glutamate[169], <strong>and</strong> to metallic compounds containing cobalt[146]. In many cases <strong>the</strong>re is a partial or completeresolution <strong>of</strong> symptoms following withdrawal <strong>of</strong> <strong>the</strong>provoking agent.OFG may be part <strong>of</strong> a spectrum <strong>of</strong> diseases that includesMelkersson Rosenthal syndrome (MRS) <strong>and</strong>cheilitis granulomatosa ( Miescher’s syndrome). MRS,in its complete form, is a triad <strong>of</strong> fissured tongue, labialor facial swelling due to granulomatous inflammation,<strong>and</strong> facial nerve palsy, which may be <strong>the</strong> first indication<strong>of</strong> <strong>the</strong> disease [192]. Cheilitis granulomatosa is probablymerely an isolated manifestation <strong>of</strong> OFG.3.6.6 Chronic Marginal Gingivitis<strong>and</strong> Localised GingivalFibrous HyperplasiaChronic marginal gingivitis <strong>of</strong> variable degree is socommon as to be almost universal. It represents a response<strong>of</strong> <strong>the</strong> gingival tissues to accumulation <strong>of</strong> dentalmicrobial plaque around <strong>the</strong> teeth. If left untreated <strong>the</strong>inflammation can become more severe <strong>and</strong> extend into<strong>the</strong> underlying periodontal tissues causing loss <strong>of</strong> periodontalligament attachment <strong>and</strong> a pocket develops between<strong>the</strong> tooth <strong>and</strong> <strong>the</strong> overlying gingiva exacerbating<strong>the</strong> tendency for plaque accumulation. Eventually, <strong>the</strong>reis progressive resorption <strong>of</strong> <strong>the</strong> supporting alveolar boneleading to loosening or loss <strong>of</strong> <strong>the</strong> tooth.Chronic marginal gingivitis is characterised microscopicallyby mild vascular hyperaemia <strong>and</strong> densechronic inflammatory infiltration. The crevicular epi<strong>the</strong>liumis ulcerated <strong>and</strong> may become hyperplastic withthin, irregular <strong>and</strong> anastomosing processes extendinginto <strong>the</strong> gingival connective tissue. There may be consid-


Oral Cavity Chapter 3 93erable intercellular oedema <strong>and</strong> infiltration <strong>of</strong> <strong>the</strong> spongiformspaces by neutrophils, especially in <strong>the</strong> presence<strong>of</strong> gross dental plaque <strong>and</strong> calculus deposits. Many lymphocytes<strong>and</strong> plasma cells are present in <strong>the</strong> inflammatoryinfiltrate <strong>and</strong> dense, basophilic, granular deposits<strong>of</strong> extracellular immunoglobulin are common. Russellbodies may be a conspicuous feature. There is variableloss <strong>of</strong> collagen in areas <strong>of</strong> severe inflammation. However,in younger patients especially, <strong>the</strong>re may be a proliferativeresponse with extensive formation <strong>of</strong> new fibroustissue leading to localised areas <strong>of</strong> fibrous hyperplasia.The enlarged gingiva may prevent effective cleaning <strong>of</strong><strong>the</strong> related tooth, predisposing to fur<strong>the</strong>r plaque accumulation<strong>and</strong> progressive inflammation. This type <strong>of</strong> localisedinflammatory gingival hyperplasia is seen muchmore frequently on <strong>the</strong> buccal or labial aspects <strong>of</strong> <strong>the</strong>gingiva than in <strong>the</strong> palatal or lingual areas. Such overgrowths,although frequently removed as part <strong>of</strong> a gingivectomyprocedure are not commonly sent for histologicalexamination.3.6.7 Peripheral Giant Cell Granuloma(Giant Cell Epulis)This lesion is found on <strong>the</strong> gingiva or edentulous alveolus<strong>and</strong> is thought to originate from elements <strong>of</strong> <strong>the</strong> periodontalligament. It is seen across a wide age range, but<strong>the</strong> peak incidence is between 30 <strong>and</strong> 50 years. Lesionstend to affect <strong>the</strong> area anterior to <strong>the</strong> permanent molars<strong>and</strong> are slightly more frequent in <strong>the</strong> m<strong>and</strong>ible. The giantcell epulis usually forms a fleshy, bluish swelling thatmay be sessile or broadly pedunculated <strong>and</strong> <strong>the</strong> surfaceis <strong>of</strong>ten ulcerated. There may be erosion <strong>of</strong> <strong>the</strong> underlyingbone or periodontium.Microscopically, <strong>the</strong>re is usually an uninvolved zone<strong>of</strong> fibrous tissue between <strong>the</strong> lesion <strong>and</strong> <strong>the</strong> overlying epi<strong>the</strong>lium,but this is lost if <strong>the</strong>re is inflammation or ulceration.The lesion consists <strong>of</strong> a matrix <strong>of</strong> plump, spindle-shapedcells with interspersed multinucleated <strong>and</strong>osteoclast-like giant cells. These can be numerous <strong>and</strong>may be confluent, blurring <strong>the</strong> distinction between eacho<strong>the</strong>r <strong>and</strong> <strong>the</strong> stromal cells. The multinucleated cellsare large <strong>and</strong> contain about 10–20 nuclei. There are twotypes: <strong>the</strong> most common have lightly eosinophilic cytoplasm<strong>and</strong> large vesicular nuclei with prominent nucleoliwhile <strong>the</strong> o<strong>the</strong>r type has much more densely stainedcytoplasm, <strong>and</strong> pyknotic <strong>and</strong> densely haematoxyphilicnuclei. The latter are probably a degenerative form <strong>of</strong><strong>the</strong> first type. The multinucleated cells are thought to beformed by fusion <strong>of</strong> bone marrow-derived mononuclearpre-osteoclasts [84, 101]. The lesion is usually very vascular<strong>and</strong> giant cells may be seen within <strong>the</strong> dilated vascularspaces. Red blood cell extravasation <strong>and</strong> haemosiderindeposition is common. Mitoses can <strong>of</strong>ten be seenin stromal <strong>and</strong> endo<strong>the</strong>lial cells, but this observation hasno bearing on <strong>the</strong> likely behaviour. Osseous metaplasia<strong>and</strong> dystrophic calcification may be present, usually in<strong>the</strong> middle or deeper aspects <strong>of</strong> <strong>the</strong> lesion.Giant cell epulis is indistinguishable microscopicallyfrom central giant cell granuloma <strong>and</strong> <strong>the</strong> brown tumour<strong>of</strong> hyperparathyroidism. Radiographs should betaken to exclude <strong>the</strong> possibility <strong>of</strong> a central bone lesion.If such a lesion is detected, hyperparathyroidism is excludedby assessing serum calcium, phosphate <strong>and</strong> alkalinephosphatase <strong>and</strong> measuring parathormone levels ifnecessary.Treatment is usually by conservative surgical excisionwith curettage <strong>of</strong> <strong>the</strong> underlying bone, but about 10% <strong>of</strong>cases recur. Some cases in children have been treated bydaily administration <strong>of</strong> calcitonin, delivered ei<strong>the</strong>r bysubcutaneous injections or nasal spray for up to 1 year.Although <strong>the</strong> treatment is protracted it appears to be effective[96].3.6.8 Pyogenic GranulomaPyogenic granulomas are most common on <strong>the</strong> gingivae<strong>and</strong> less frequently in o<strong>the</strong>r intraoral sites, particularly<strong>the</strong> lip <strong>and</strong> tongue. They form solitary, s<strong>of</strong>t, red <strong>and</strong> friablenodules that bleed readily. They frequently ulcerate<strong>and</strong> are covered by a fibrinous slough. Gingival lesionsmay be seen in pregnancy <strong>and</strong> appear to be a focal exacerbation<strong>of</strong> pregnancy gingivitis [113]. Pregnancy epulides(“pregnancy tumour”) usually manifest towards <strong>the</strong>end <strong>of</strong> <strong>the</strong> first trimester. They have a strong tendency torecur if removed before parturition <strong>and</strong> may show partialor complete spontaneous resolution if left followingdelivery. Occasionally, similar lesions are seen in o<strong>the</strong>rparts <strong>of</strong> <strong>the</strong> mouth during pregnancy, particularly <strong>the</strong>dorsum <strong>of</strong> <strong>the</strong> tongue, <strong>and</strong> <strong>the</strong>y are termed granulomagravidarum [52].Microscopically, oral pyogenic granulomas consist <strong>of</strong>numerous large, thin-walled, anastomosing blood vesselsin a loose, oedematous <strong>and</strong> moderately cellular stroma.Older lesions may show some fibrosis. Inflammationis very variable <strong>and</strong> can be minimal or absent. However,if <strong>the</strong> lesion ulcerates <strong>the</strong>re may an intense inflammatoryinfiltration. Foci <strong>of</strong> papillary endo<strong>the</strong>lial hyperplasiaare an occasional feature.Although excision is usually curative, rare cases canshow repeated recurrences <strong>and</strong> require more extensivesurgery for <strong>the</strong>ir eradication.3.6.9 Pulse (Vegetable) GranulomaThis unusual <strong>and</strong> uncommon chronic inflammatory lesion<strong>of</strong> oral tissues has been described under a variety<strong>of</strong> terms including chronic periostitis, giant-cell hyalinangiopathy, oral vegetable granuloma <strong>and</strong> hyaline ring


94 J.W. Eveson3Fig. 3.19. Pulse granuloma showing chronic inflammation <strong>and</strong>eosinophilic, hyaline rings toge<strong>the</strong>r with multinucleated foreignbody-typegiant cellsFig. 3.20. Giant cell fibroma showing stellate <strong>and</strong> angular fibroblastsin collagenous matrixgranuloma [171]. Most cases are seen in <strong>the</strong> premolar/molar region <strong>of</strong> <strong>the</strong> edentulous m<strong>and</strong>ible <strong>and</strong> <strong>the</strong> mostcommon complaints are recurrent swelling <strong>and</strong> tenderness.Fifty-three percent <strong>of</strong> cases are extraosseous (peripheral)<strong>and</strong> radiographs <strong>of</strong>ten show a poorly definederosion <strong>of</strong> <strong>the</strong> underlying alveolar bone. Intraosseous orcentral lesions (42%) show an irregular radiolucent areathat is non-diagnostic. Occasionally, <strong>the</strong> lesion is foundwithin <strong>the</strong> fibrous wall <strong>of</strong> an odontogenic or nasopalatineduct cyst [140].Microscopy shows chronic inflammation <strong>and</strong> eosinophilic,hyaline rings toge<strong>the</strong>r with multinucleatedforeign-body-type giant cells (Fig. 3.19). The ringsmay be complete or horse-shoe shaped <strong>and</strong> may enclosegiant cells, connective tissue <strong>and</strong> blood vessels.Haemosiderin within <strong>the</strong> centre <strong>of</strong> <strong>the</strong> rings is a frequentfinding. The suggestion that <strong>the</strong> histologicalappearances are due to thickening <strong>and</strong> hyalinisation<strong>of</strong> <strong>the</strong> walls <strong>of</strong> blood vessels is not supported by mostobservers. Light <strong>and</strong> electron microscopical findingssuggest that <strong>the</strong> rings are <strong>the</strong> cell walls <strong>of</strong> vegetableremains, <strong>of</strong>ten with collagen attached to <strong>the</strong>ir surface[71]. There does not appear to be any evidence that<strong>the</strong>se appearances are exclusively due to pulses. Completeexcision is curative.3.7 Benign Tumours<strong>and</strong> Pseudotumours3.7.1 Giant Cell FibromaGiant cell fibroma is an unusual but distinctive type<strong>of</strong> fibrous overgrowth. It is typically less than 5 mm indiameter <strong>and</strong> usually presents as a pedunculated polypwith a lobulated surface. Most are seen in <strong>the</strong> first threedecades <strong>of</strong> life. Although <strong>the</strong>y can form anywhere in <strong>the</strong>oral mucosa, about half <strong>of</strong> cases are seen on <strong>the</strong> gingiva[79].Microscopically, <strong>the</strong>y consist <strong>of</strong> interweaving bundles<strong>of</strong> collagenous connective tissue with a prominent capillarynetwork that surround stellate or angular fibroblasticgiant cells with large vesicular nuclei (Fig. 3.20). Occasionalcells may have several nuclei. These cells mayhave conspicuous dendritic processes <strong>and</strong> some containmelanin pigment. The giant cells are positive for vimentin,but negative for S-100, cytokeratin, leukocyte commonantigen <strong>and</strong> neur<strong>of</strong>ilament [107]. Conservative surgicalexcision is usually curative.3.7.2 Lingual ThyroidEctopic lingual thyroid is a rare developmental anomalydue to failure <strong>of</strong> <strong>the</strong> thyroglossal duct to migrate caudallyfrom <strong>the</strong> foramen caecum [13]. It is seen in femalesabout four times more frequently than males <strong>and</strong> usuallypresents in middle age. The lingual gl<strong>and</strong> is seen in<strong>the</strong> base <strong>of</strong> <strong>the</strong> tongue, deep to <strong>the</strong> foramen caecum. It is<strong>of</strong>ten asymptomatic, but may cause dysphagia, dysphoniaor dyspnoea. Symptoms may coincide with puberty,pregnancy or <strong>the</strong> menopause due to hyperplasia secondaryto raised levels <strong>of</strong> thyroid-stimulating hormone. Inaddition, any <strong>of</strong> <strong>the</strong> diseases involving <strong>the</strong> conventionalthyroid gl<strong>and</strong>, including inflammatory conditions, adenomas<strong>and</strong> carcinomas, can affect <strong>the</strong> ectopic thyroidtissue.Microscopy typically shows normal thyroid tissue.As many as 70% <strong>of</strong> patients with lingual thyroid haveno o<strong>the</strong>r thyroid tissue present, so it is essential that presurgicalevaluation includes appropriate imaging <strong>and</strong>assessment <strong>of</strong> function using 131I or 99mTc pertechnetate.


Oral Cavity Chapter 3 953.7.3 Verruciform XanthomaThis rare but distinctive lesion, first described in 1971[166], forms most commonly in <strong>the</strong> oral cavity [82,135]. Extraoral locations include <strong>the</strong> male <strong>and</strong> femalegenitalia. There is no association with HPV in <strong>the</strong> vastmajority <strong>of</strong> cases studied. Verruciform xanthomas areseen at all ages, but are most frequent in <strong>the</strong> fifth toseventh decades. Incidence is roughly equal between<strong>the</strong> sexes. They usually present as solitary, painless,discrete nodules that may be <strong>the</strong> colour <strong>of</strong> <strong>the</strong> surroundingmucosa, reddish or pink. They can be sessileor pedunculated <strong>and</strong> <strong>the</strong> surface can be domed or flat,<strong>and</strong> can be keratotic or papilliferous. They usually havesharply defined margins <strong>and</strong> are typically less than acentimetre in diameter. They do not appear to be relatedto any local irritating factors <strong>and</strong> most cases areasymptomatic. The gingival margin accounts for 85%<strong>of</strong> cases. O<strong>the</strong>r common sites include <strong>the</strong> hard palate,tongue, buccal mucosa <strong>and</strong> a variety <strong>of</strong> o<strong>the</strong>r intraoralsites.Microscopy typically shows corrugated, hyperplasticepi<strong>the</strong>lium with elongated, broad rete ridges thatextend to a straight, well-defined lower border [127].There are deep clefts within <strong>the</strong> epi<strong>the</strong>lium that <strong>of</strong>tencontain keratinised plugs. The surface shows parakeratinisedspikes, which <strong>of</strong>ten stain a deep orange colour.There may be secondary c<strong>and</strong>idal infestation <strong>of</strong><strong>the</strong> surface keratin layers [125]. The characteristic featureis <strong>the</strong> presence <strong>of</strong> vacuolated, foamy xanthomatouscells, which fill <strong>the</strong> papillary corium. These xanthomacells can occasionally extend into <strong>the</strong> overlyingepi<strong>the</strong>lium or into <strong>the</strong> deeper corium. The xanthomacells have been shown to be derived from cells <strong>of</strong>monocyte/macrophage lineage [121].The lesions are entirely benign <strong>and</strong> recurrence followingeven conservative surgery is very rare.3.7.4 HaemangiomasICD-O:9120/0Haemangiomas are vas<strong>of</strong>ormative tumours that closelyresemble normal vessels. They are amongst <strong>the</strong> mostcommon s<strong>of</strong>t tissue tumours <strong>and</strong> about a third <strong>of</strong> allcases involve <strong>the</strong> head <strong>and</strong> neck region. In this locationcongenital or neonatal lesions are relatively uncommon<strong>and</strong> tend to involve <strong>the</strong> lips <strong>and</strong> parotid gl<strong>and</strong>s. The majority<strong>of</strong> cases are seen in older individuals, <strong>and</strong> <strong>the</strong> mostcommon sites are <strong>the</strong> lips <strong>and</strong> post-commissural buccalmucosa <strong>and</strong> <strong>the</strong> lateral border <strong>of</strong> <strong>the</strong> tongue. There is amale predominance <strong>of</strong> about 2:1. Haemangiomas typicallyform painless, flat or nodular, s<strong>of</strong>t <strong>and</strong> purplishredlesions. They are usually well circumscribed <strong>and</strong>may blanch on pressure. Congenital lesions behave likehamartomas <strong>and</strong> increase in size in proportion to generalsomatic growth <strong>and</strong> tend to stabilise in size in earlyadult life. Those presenting in older individuals mayshow slow but progressive growth over several years.Their classification <strong>and</strong> microscopic aspects are discussedin <strong>the</strong> section on s<strong>of</strong>t tissue tumours in Chap. 2[3, 48, 53, 170, 175, 181].3.7.5 LymphangiomaICD-O:9170/0Lymphangioma is a benign, cavernous or cystic vascularlesion composed <strong>of</strong> dilated lymphatic channels.The head <strong>and</strong> neck region is <strong>the</strong> most common site <strong>of</strong>involvement. Many lesions are present at birth or developwithin <strong>the</strong> first few years <strong>of</strong> life. There appearsto be no sex predilection. They usually form painlessswellings that are frequently superficial, but some extenddeeply into <strong>the</strong> surrounding tissues <strong>and</strong> are illdefined.Oral lesions are seen most commonly in <strong>the</strong> tongue<strong>and</strong> lips where <strong>the</strong>y may cause macroglossia <strong>and</strong> macrocheiliarespectively. Involvement <strong>of</strong> underlying bonein oral lesions is uncommon [133]. Mucosal lymphangiomasusually have a pale, translucent surface, whichis nodular or bossellated. It is common for black areasto appear in <strong>the</strong> lesion due to focal areas <strong>of</strong> haemorrhage.In <strong>the</strong> neck, <strong>the</strong> lesions tend to be larger <strong>and</strong> showmore extensive cystic dilatation. These lymphangiomasare <strong>the</strong>n frequently called cystic lymphangiomas or cystichygromas [91]. Some are associated with Turner syndrome(Monosomy X) [28]. The most common locationsfor cystic hygroma are <strong>the</strong> posterior triangle, subm<strong>and</strong>ibularregion <strong>and</strong> floor <strong>of</strong> <strong>the</strong> mouth. It can extend upwardsto involve <strong>the</strong> cheek <strong>and</strong> parotid gl<strong>and</strong>, forwardsinto <strong>the</strong> anterior triangle or downwards into <strong>the</strong> mediastinum.Some cystic hygromas are severely disfiguring<strong>and</strong> <strong>the</strong>y can compromise swallowing or breathing. Theclassification <strong>and</strong> microscopic aspects <strong>of</strong> oral lymphangiomasare discussed in Chap. 9.3.7.6 Benign Nerve Sheath TumoursNeur<strong>of</strong>ibroma <strong>and</strong> schwannoma are <strong>the</strong> two mostcommon benign tumours <strong>of</strong> nerve sheath origin. Although<strong>the</strong>y both appear to be derived from Schwanncells <strong>the</strong>y have distinctive clinical <strong>and</strong> microscopicalfeatures.


96 J.W. Eveson33.7.6.1 Neur<strong>of</strong>ibromaICD-O:9540/0Solitary neur<strong>of</strong>ibromas are relatively uncommon in<strong>the</strong> or<strong>of</strong>acial region <strong>and</strong> tend to affect people in <strong>the</strong>20–40 years age group [189]. Most tumours are sporadic,but <strong>the</strong> possibility <strong>of</strong> neur<strong>of</strong>ibromatosis type 1should always be considered when dealing with <strong>the</strong>selesions. They usually form small, painless, expansilesubmucosal nodules. The tongue is <strong>the</strong> most commonintraoral site, but occasionally <strong>the</strong>y develop on<strong>the</strong> inferior dental nerve <strong>and</strong> appear as a fusiform radiolucentarea along <strong>the</strong> course <strong>of</strong> <strong>the</strong> inferior dentalcanal.3.7.6.2 SchwannomaICD-O:9560/0Schwannoma ( neurilemmoma; neurinoma) is a benignneoplasm <strong>of</strong> Schwann cell origin. Patients are usuallyin <strong>the</strong> third or fourth decades <strong>and</strong> <strong>the</strong> tumour is morecommon in women than men. Lesions in <strong>the</strong> mouthtend to form small, painless <strong>and</strong> slow-growing swellings<strong>and</strong> <strong>the</strong> tongue is <strong>the</strong> most common site. However,occasional cases can be several centimetres in diameter[41, 87].3.7.6.3 Neur<strong>of</strong>ibromatosisICD-O:9540/0Von Recklinghausen disease or neur<strong>of</strong>ibromatosis type Iis inherited as an autosomal dominant trait <strong>and</strong> <strong>the</strong> generesponsible is located on chromosome 17 [153]. It is relativelycommon <strong>and</strong> affects about 1 in 4,000 births. It isusually diagnosed before <strong>the</strong> age <strong>of</strong> 10 years because <strong>of</strong><strong>the</strong> characteristic cutaneous lesions <strong>and</strong> <strong>the</strong> frequentfamily history.Clinically, it is characterised by cutaneous neur<strong>of</strong>ibromasthat are usually associated with café au lait pigmentation<strong>of</strong> <strong>the</strong> skin. Lesions may be focal, but sometimes<strong>the</strong>re can be thous<strong>and</strong>s <strong>of</strong> tumours <strong>and</strong> <strong>the</strong> conditionis <strong>the</strong>n grossly disfiguring. The lesions are usuallypainless, but itching can be a serious problem. Therecan be overgrowth <strong>of</strong> bone <strong>and</strong> associated s<strong>of</strong>t tissueleading to bizarre localised gigantism. Nearly a quarter<strong>of</strong> cases involve <strong>the</strong> head <strong>and</strong> neck, but only about 5% affect<strong>the</strong> oral cavity.3.7.6.4 Multiple Neuromasin Endocrine NeoplasiaSyndromeICD-O:9570/0Multiple endocrine neoplasia syndrome type 2B is anautosomal dominant condition characterised by <strong>the</strong>presence <strong>of</strong> mucosal neuromas toge<strong>the</strong>r with medullarycarcinoma <strong>of</strong> <strong>the</strong> thyroid gl<strong>and</strong> <strong>and</strong> phaeochromocytoma[147]. Nearly 90% <strong>of</strong> patients with <strong>the</strong> conditionhave point mutations at codon 918 <strong>of</strong> <strong>the</strong> RET proto-oncogene.Clinically, patients <strong>of</strong>ten have a Marfanoid habituswith arachnodactyly <strong>and</strong> a narrow face. Mucosal neuromasare <strong>the</strong> most consistent feature <strong>of</strong> <strong>the</strong> disease <strong>and</strong>may be pathognomonic. They tend to form on <strong>the</strong> lateralmargins <strong>and</strong> dorsum <strong>of</strong> <strong>the</strong> tongue <strong>and</strong> appear asmultiple, small, painless nodules. These nodules may be<strong>the</strong> first indication <strong>of</strong> <strong>the</strong> condition. The lips are sometimesenlarged <strong>and</strong> blubbery. Mucosal neuromas <strong>of</strong>tenaffect <strong>the</strong> palpebral conjunctiva <strong>and</strong> can also involve <strong>the</strong>sclera.Microscopy shows a partially encapsulated tangledmass <strong>of</strong> small nerve fibres, <strong>of</strong>ten with a thickened perineurium.These nerves lie in a loose fibrous stroma. Thenerves usually stain with S-100 <strong>and</strong> may stain with epi<strong>the</strong>lialmembrane antigen indicating perineurial differentiation.The mucosal neuromas <strong>the</strong>mselves rarely cause clinicalproblems, but may act as indicators <strong>of</strong> <strong>the</strong> more seriousaspects <strong>of</strong> <strong>the</strong> syndrome.3.7.7 Granular Cell Tumour(Granular Cell Myoblastoma)ICD-O:9580/0Granular cell tumours are uncommon, but about half <strong>of</strong>all cases involve <strong>the</strong> head <strong>and</strong> neck region. The tongue,particularly <strong>the</strong> dorsum, is <strong>the</strong> most common site. Thepeak incidence is in middle life <strong>and</strong> 10–20% <strong>of</strong> cases aremultiple. There is a female predominance <strong>of</strong> about 2:1.They usually form nondescript, painless swellings, butoccasionally surface c<strong>and</strong>idal infestation causes <strong>the</strong> lesionto present as a white plaque [59, 108]. See for thislesion also Chap. 7.3.8 Squamous Cell Carcinoma3.8.1 IntroductionSquamous cell carcinomas account for about 90% <strong>of</strong>all malignant neoplasms in <strong>the</strong> mouth <strong>and</strong> orophar-


Oral Cavity Chapter 3 97ynx. It is important to consider <strong>the</strong> site <strong>of</strong> involvementas <strong>the</strong> epidemiological factors can vary considerably intumours in different intraoral locations. There is typicallya higher frequency in men than women, <strong>and</strong> thisis attributed to <strong>the</strong> use <strong>of</strong> tobacco <strong>and</strong> alcohol [17]. Ithas been estimated that as many as 75% <strong>of</strong> cases <strong>of</strong> oralsquamous cell carcinomas in Western countries <strong>and</strong> Japancan be ascribed to <strong>the</strong>se factors. Globally, oral canceraccounts for 5% <strong>of</strong> all malignancies in men <strong>and</strong> 2%in women [134]. Much higher rates, however, are seen inboth men <strong>and</strong> women in parts <strong>of</strong> sou<strong>the</strong>ast Asia, where<strong>the</strong>y are usually associated with <strong>the</strong> habitual use <strong>of</strong> arecanut <strong>and</strong> tobacco products.3.8.2 Clinical FeaturesDespite <strong>the</strong> fact that oral tumours frequently causesymptoms, <strong>and</strong> <strong>the</strong> mouth can be readily visualisedwith simple equipment, many oral cancers present at arelatively advanced stage where treatment may be disfiguring<strong>and</strong> prognosis is poor. This is <strong>of</strong>ten becausemany patients are elderly <strong>and</strong> frail <strong>and</strong> frequently weardental pros<strong>the</strong>ses <strong>and</strong> are accustomed to minor degrees<strong>of</strong> oral discomfort. In addition, early lesions may not beregarded as suspicious by <strong>the</strong> patient or <strong>the</strong> clinician <strong>and</strong>may <strong>the</strong>refore be treated empirically with antibacterialor antifungal preparations.Any part <strong>of</strong> <strong>the</strong> oral mucosa can be <strong>the</strong> site <strong>of</strong> development<strong>of</strong> squamous cell carcinomas. The commonoral locations can show wide variations in different geographicalareas depending on <strong>the</strong> prevalent risk factors.The intraoral subsites include <strong>the</strong> buccal mucosa,tongue, floor <strong>of</strong> mouth, upper <strong>and</strong> lower gingivae <strong>and</strong>alveolar processes, <strong>the</strong> hard palate <strong>and</strong> retromolar trigone.As <strong>the</strong> clinical presentation can vary according to<strong>the</strong> specific sites <strong>of</strong> involvement, <strong>the</strong>se will be discussedseparately.3.8.2.1 Buccal MucosaThe buccal mucosa extends from <strong>the</strong> commissureanteriorly to <strong>the</strong> retromolar trigone posteriorly <strong>and</strong>from <strong>the</strong> upper <strong>and</strong> lower vestibular reflections. Themajority <strong>of</strong> carcinomas arise from <strong>the</strong> posterior areawhere <strong>the</strong>y are commonly traumatised by <strong>the</strong> molarteeth. They soon spread into <strong>the</strong> underlying buccinatormuscle <strong>and</strong> though insidious initially <strong>the</strong>y may eventuallycause trismus. Bone, however, is generally involvedonly in advanced tumours. Tumours at this site <strong>of</strong>tenextend posteriorly into <strong>the</strong> palatoglossal fold <strong>and</strong> tonsillarfossa. Metastases are most common in <strong>the</strong> subm<strong>and</strong>ibular,submental, parotid <strong>and</strong> lateral pharyngeallymph nodes.3.8.2.2 TongueThe tongue is <strong>the</strong> most common oral location <strong>of</strong> squamouscell carcinoma <strong>and</strong> can account for half <strong>of</strong> allcases. The majority affect <strong>the</strong> middle third <strong>of</strong> <strong>the</strong> lateralborder <strong>and</strong> adjacent ventral surface. The dorsum is avery uncommon site <strong>and</strong> tumours arising <strong>the</strong>re may beassociated with precursor lesions such as lichen planus<strong>and</strong> c<strong>and</strong>idal leukoplakia. Lingual tumours are <strong>of</strong>tenexophytic <strong>and</strong> ulceration is common. Even clinicallysmall tumours can infiltrate deeply into <strong>the</strong> underlyingmuscle. With progressive growth tumours becomeindurated <strong>and</strong> frequently develop characteristic rolled,raised, everted margins. Infiltration <strong>of</strong> <strong>the</strong> lingual musculaturemay cause pain, dysphagia <strong>and</strong> dysphonia.Half <strong>of</strong> patients have regional lymph node metastasesat presentation. Tumours towards <strong>the</strong> tip <strong>of</strong> <strong>the</strong> tonguedrain to <strong>the</strong> submental <strong>and</strong> <strong>the</strong>nce to <strong>the</strong> jugulo-digastriclymph node, <strong>and</strong> those located on <strong>the</strong> dorsum <strong>and</strong>lateral borders tend to involve <strong>the</strong> subm<strong>and</strong>ibular <strong>and</strong>jugulo-digastric nodes. Contralateral or bilateral spreadis relatively common, particularly in tumours arisinganteriorly.3.8.2.3 Floor <strong>of</strong> MouthThe floor <strong>of</strong> <strong>the</strong> mouth is a horseshoe-shaped mucosaltrough extending between <strong>the</strong> lower lingual alveolarmucosa <strong>and</strong> <strong>the</strong> ventral lingual mucosa. It is<strong>the</strong> second most common site for intraoral squamouscell carcinomas <strong>and</strong> shows <strong>the</strong> highest frequency <strong>of</strong>small <strong>and</strong> symptomless tumours [115]. Tumours aremost frequent in <strong>the</strong> anterior segment <strong>and</strong> tumours<strong>the</strong>re tend to spread superficially ra<strong>the</strong>r than deeply.Involvement <strong>of</strong> <strong>the</strong> subm<strong>and</strong>ibular duct can causeobstructive sialadenitis <strong>and</strong> tumours can also extenddown <strong>the</strong> duct itself. If <strong>the</strong> tumour extends to involve<strong>the</strong> m<strong>and</strong>ible, <strong>the</strong>re can be spread along <strong>the</strong> periodontalligament <strong>and</strong> subperiostally [106]. Lymphatic involvementis early but less frequent than tumours <strong>of</strong><strong>the</strong> tongue itself.3.8.2.4 Gingiva<strong>and</strong> Alveolar RidgeTumours at this site can be exophytic resembling dentalabscesses or epulides, or ulcerated <strong>and</strong> fixed to <strong>the</strong>underlying bone. They account for about 20% <strong>of</strong> oraltumours. In parts <strong>of</strong> <strong>the</strong> USA <strong>the</strong>re is a very high frequencyin women who practise snuff dipping. Relatedteeth are <strong>of</strong>ten loosened <strong>and</strong> <strong>the</strong>re is extension along <strong>the</strong>periodontal ligament. On <strong>the</strong> alveolus tumours can re-


98 J.W. Eveson3semble simple lesions like denture-induced hyperplasiaor denture-related ulceration. The underlying bone maybe eroded or invaded in 50% <strong>of</strong> patients <strong>and</strong> regionalmetastases are seen in over half <strong>the</strong> patients at presentation.3.8.2.5 Hard PalateThis is a relatively uncommon site <strong>of</strong> involvement exceptin areas where reverse smoking is common [148].Tumours at this site can be exophytic or ulcerative, buttend to spread superficially ra<strong>the</strong>r than deeply.3.8.2.6 Retromolar TrigoneTumours from this site spread to <strong>the</strong> buccal mucosa laterally<strong>and</strong> distally involve <strong>the</strong> tonsillar area. They canpenetrate into <strong>the</strong> parapharyngeal area <strong>and</strong> may showextensive spread along <strong>the</strong> lingual <strong>and</strong> inferior alveolarnerves. In addition, tumours frequently erode or invade<strong>the</strong> adjacent m<strong>and</strong>ible.3.8.3 StagingStaging <strong>of</strong> oral squamous cell carcinomas is undertakenusing <strong>the</strong> current TNM Classification [67, 167].References1. Abdollahi M, Radfar MA (2003) A review <strong>of</strong> drug-inducedoral reactions. J Contemp Dent Pract 4:10–312. Amagai M, Karpati S, Prussick R, Klaus-Kovtun V, StanleyJR (1992) Autoantibodies against <strong>the</strong> amino-terminal cadherin-likebinding domain <strong>of</strong> pemphigus vulgaris antigenare pathogenic. J Clin Investig 90:919–9263. Amerigo J, Matilla A, Gonzalez-Campora R, Galera-DavidsonH (1979) Vegetant intravascular hemangioendo<strong>the</strong>lioma<strong>of</strong> <strong>the</strong> skin. Report <strong>of</strong> 3 cases with a critical pathogeneticstudy. Dermatologica 159:476–4814. Andreasen JO (1968) Oral lichen planus. II. A histologic evaluation<strong>of</strong> 97 cases. Oral Surg Oral Med Oral Pathol 25:158–1665. Anhalt GJ, Kim SC, Stanley JR, Korman NJ et al. (1990) Paraneoplasticpemphigus. An autoimmune mucocutaneous diseaseassociated with neoplasia. N Engl J Med 320:1729–17356. Assimakopoulos D, Patrikakos G, Fotika C, Elisaf M (2002)Benign migratory glossitis or geographic tongue: an enigmaticoral lesion. Am J Med 113:751–7557. Aurelian L, Ono F, Burnett J (2003) Herpes simplex virus(HSV)-associated ery<strong>the</strong>ma multiforme (HAEM): a viraldisease with an autoimmune component. Dermatol OnlineJ 9:18. Ayangco L, Rogers RS III (2003) Oral manifestations <strong>of</strong> ery<strong>the</strong>mamultiforme. Dermatol Clin 21:195–2059. Barker BF, Carpenter WM, Daniels et al. (1997) Oral mucosalmelanomas: <strong>the</strong> WESTOP Banff workshop proceedings.Western Society <strong>of</strong> Teachers <strong>of</strong> Oral <strong>Pathology</strong>. Oral SurgOral Med Oral Pathol Oral Radiol Endod 83:672–67910. Barrett AW, Bennett JH, Speight PM (1995) A clinicopathological<strong>and</strong> immunohistochemical analysis <strong>of</strong> primary oralmucosal melanoma. Eur J Cancer B Oral Oncol 31B:100–10511. Barrett AW, Porter SR, Scully C, Eveson JW, Griffiths MJ(1994) Oral melanotic macules that develop after radiation<strong>the</strong>rapy. Oral Surg Oral Med Oral Pathol 77:431–43412. Bastiaens MT, Zwan NV, Verschueren GL, Sto<strong>of</strong> TJ, NieboerC (1994) Three cases <strong>of</strong> pemphigus vegetans: induction byenalapril – association with internal malignancy. Int J Dermatol33:168–17113. Batsakis JG, El-Naggar AK, Luna MA (1996) Thyroid gl<strong>and</strong>ectopias. Ann Otol Rhinol Laryngol 105:996–100014. Batsakis JG, Suarez P (2000) Mucosal melanomas: a review.Adv Anat Pathol 7:167–18015. Becker BA, Gaspari AA (1993) Pemphigus vulgaris <strong>and</strong> vegetans.Dermatol Clin 11:429–45216. Benateau H, Rigau V, Comoz F, Benchemam Y, Galateau F,Compere JF (2003) Tumour <strong>of</strong> <strong>the</strong> juxtaoral organ. Int J OralMaxill<strong>of</strong>ac Surg 32:101–10317. Blot WJ, McLaughlin JK, Winn DM et al. (1988) Smoking<strong>and</strong> drinking in relation to oral <strong>and</strong> pharyngeal cancer. CancerRes 48:3282–328718. Bonen DK, Cho JH (2003) The genetics <strong>of</strong> inflammatorybowel disease. Gastroenterology 124:521–53619. Bozkurt T, Langer M, Fendel K, Lux G (1992) Granulomatoustonsillitis. A rare extraintestinal manifestation <strong>of</strong> Crohn’sdisease. Dig Dis Sci 37:1127–113020. Bozzo L, Machado MA, de Almeida OP, Lopes MA, ColettaRD (2000) Hereditary gingival fibromatosis: report <strong>of</strong> threecases. J Clin Pediatr Dent 25:41–4621. Brooke AE, Eveson JW, Luker J, Oakhill A (1999) Oral presentation<strong>of</strong> a novel variant <strong>of</strong> herpes simplex infection in agroup <strong>of</strong> bone marrow transplant patients: a report <strong>of</strong> fivecases. Br J Dermatol 141:381–36322. Buchner A, Hansen LS (1979) Melanotic macule <strong>of</strong> <strong>the</strong> oralmucosa. A clinicopathologic study <strong>of</strong> 105 cases. Oral SurgOral Med Oral Pathol 48:244–24923. Buchner A, Hansen LS (1980) Amalgam pigmentation (amalgamtattoo) <strong>of</strong> <strong>the</strong> oral mucosa. A clinicopathologic study <strong>of</strong>268 cases. Oral Surg Oral Med Oral Pathol 49:139–14724. Buchner A, Leider AS, Merrell PW, Carpenter WM (1990)Melanocytic nevi <strong>of</strong> <strong>the</strong> oral mucosa: a clinicopathologicstudy <strong>of</strong> 130 cases from nor<strong>the</strong>rn California. J Oral PatholMed 19:197–20125. Buchner A, Lozada F, Silverman S Jr (1980) Histopathologicspectrum <strong>of</strong> oral ery<strong>the</strong>ma multiforme. Oral Surg Oral MedOral Pathol 49:221–22826. Budtz-Jorgensen E (1990) Histopathology, immunology, <strong>and</strong>serology <strong>of</strong> oral yeast infections. Diagnosis <strong>of</strong> oral c<strong>and</strong>idosis.Acta Odontol Sc<strong>and</strong> 8:37–4327. Burrows NP, Lockwood CM (1995) Antineutrophil cytoplasmicantibodies <strong>and</strong> <strong>the</strong>ir relevance to <strong>the</strong> dermatologist. Br JDermatol 132:173–18128. Byrne J, Blanc WA, Warburton D, Wigger J (1984) The significance<strong>of</strong> cystic hygroma in fetuses. Hum Pathol 15:61–6729. Card T, Logan RF, Rodrigues LC, Wheeler JG (2004) Antibioticuse <strong>and</strong> <strong>the</strong> development <strong>of</strong> Crohn’s disease. Gut 53:246–25030. Cawson RA, Lehner T (1968) Chronic hyperplastic c<strong>and</strong>idiasis– c<strong>and</strong>idal leukoplakia. Br J Dermatol 80:9–1631. Cawson RA, Rajasingham KC (1972) Ultrastructural features<strong>of</strong> <strong>the</strong> invasive phase <strong>of</strong> C<strong>and</strong>ida albicans. Br J Dermatol87:435–44332. Chan LS (2001) Mucous membrane pemphigoid. Clin Dermatol19:703–71133. Chan LS, Ahmed AR, Anhalt GJ et al. (2002) The first internationalconsensus on mucous membrane pemphigoid:definition, diagnostic criteria, pathogenic factors, medicaltreatment, <strong>and</strong> prognostic indicators. Arch Dermatol 2002138:370–37934. Chao SC, Tsai YM, Yang MH, Lee JY (2003) A novel mutationin <strong>the</strong> keratin 4 gene causing white sponge naevus. Br JDermatol 148:1125–1128


Oral Cavity Chapter 3 9935. Chaudhry AP, Hampel A, Gorlin RJ (1958) Primary malignantmelanoma <strong>of</strong> <strong>the</strong> oral cavity: a review <strong>of</strong> 105 cases. Cancer11:923–92836. Chong CY, Chan KP, Shah VA et al. (2003) H<strong>and</strong>, foot <strong>and</strong>mouth disease in Singapore: a comparison <strong>of</strong> fatal <strong>and</strong> nonfatalcases. Acta Paediatr 92:1163–116937. Cooke BED (1956) Leukoplakia buccalis <strong>and</strong> oral epi<strong>the</strong>lialnaevi: a clinical <strong>and</strong> histological study. Br Dent J 68:151–17438. Csernok E (2003) Anti-neutrophil cytoplasmic antibodies<strong>and</strong> pathogenesis <strong>of</strong> small vessel vasculitides. AutoimmunRev 2:158–16439. Daley T (1993) <strong>Pathology</strong> <strong>of</strong> intraoral sebaceous gl<strong>and</strong>s. JOral Pathol Med 22:241–24540. Daniels TE, Hansen LS, Greenspan JS et al. (1992) Histopathology<strong>of</strong> smokeless tobacco lesions in pr<strong>of</strong>essional baseballplayers. Associations with different types <strong>of</strong> tobacco. OralSurg Oral Med Oral Pathol 73:720–72541. Dayan D, Buchner A, Hirschberg A (1989) Ancient neurilemmoma(Schwannoma) <strong>of</strong> <strong>the</strong> oral cavity. J Craniomaxill<strong>of</strong>acSurg 17:280–28242. Devaney KO, Travis WD, H<strong>of</strong>fman G et al. (1990) Interpretation<strong>of</strong> head <strong>and</strong> neck biopsies in Wegener’s granulomatosis.A pathologic study <strong>of</strong> 126 biopsies in 70 patients. Am J SurgPathol 14:555–56443. Doyle JL, Geary W, Baden E (1989) Eosinophilic ulcer. J OralMaxill<strong>of</strong>ac Surg 47:349–35244. Eley BM (1982) Tissue reactions to implanted dental amalgam,including assessment by energy dispersive x-ray microanalysis.J Pathol 138:251–27245. El-M<strong>of</strong>ty SK, Swanson PE, Wick MR, Miller AS (1993) Eosinophiliculcer <strong>of</strong> <strong>the</strong> oral mucosa. Report <strong>of</strong> 38 new caseswith immunohistochemical observations. Oral Surg OralMed Oral Pathol 75:716–72246. Elzay RP (1983) Traumatic ulcerative granuloma with stromaleosinophilia (Riga-Fede’s disease <strong>and</strong> traumatic eosinophilicgranuloma). Oral Surg Oral Med Oral Pathol 55:497–50647. Eversole LR, Leider AS (1978) Maxillary intraosseous neuroepi<strong>the</strong>lialstructures resembling those seen in <strong>the</strong> organ <strong>of</strong>Chievitz. Oral Surg Oral Med Oral Pathol 46:555–55848. Eveson JW, Lucas RB (1979) Angiolymphoid hyperplasiawith eosinophilia. J Oral Pathol 8:103–10849. Fatahzadeh M, Sirois DA (2002) Multiple intraoral melanoacanthomas:a case report with unusual findings. Oral SurgOral Med Oral Pathol Oral Radiol Endod 94:54–5650. Fauci AS, Wolff SM (1973) Wegener’s granulomatosis: studiesin eighteen patients <strong>and</strong> a review <strong>of</strong> <strong>the</strong> literature. Medicine(Baltimore) 52:535–56151. Femiano F, Scully C, Gombos F (2003) Linear IgA dermatosisinduced by a new angiotensin-converting enzyme inhibitor.Oral Surg Oral Med Oral Pathol Oral Radiol Endod95:169–17352. Fenton JE, Timon CI, McShane DP (1996) Lingual granulomagravidarum. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 114:682–68353. Fetsch JF (2002) Endo<strong>the</strong>lioid haemangioma. In: FletcherCDM, Unni KK, Mertens F (eds) <strong>Pathology</strong> <strong>and</strong> genetics <strong>of</strong>tumours <strong>of</strong> s<strong>of</strong>t tissue <strong>and</strong> bone. WHO Classification <strong>of</strong> Tumours.IARC Press, Lyon, pp 159–16054. Ficarra G, Prignano F, Romagnoli P (1997) Traumatic eosinophilicgranuloma <strong>of</strong> <strong>the</strong> oral mucosa: a CD30+(Ki-1) lymphoproliferativedisorder? Oral Oncol 33:375–37955. Ficarra G, Shillitoe EJ, Adler-Storthz K et al. (1990) Oral melanoticmacules in patients infected with human immunodeficiencyvirus. Oral Surg Oral Med Oral Pathol 70:748–75556. Field EA, Allan RB (2003) Review article: oral ulceration –aetiopathogenesis, clinical diagnosis <strong>and</strong> management in <strong>the</strong>gastrointestinal clinic. Aliment Pharmacol Ther 18:949–96257. Field EA, Tyldesley WR (1989) Oral Crohn’s disease revisited– a 10-year-review. Br J Oral Maxill<strong>of</strong>ac Surg 27:114–12358. Fienberg R (1981) The protracted superficial phenomenon inpa<strong>the</strong>rgic (Wegener’s) granulomatosis. Hum Pathol 12:458–46759. Fine SW, Li M (2003) Expression <strong>of</strong> calretinin <strong>and</strong> <strong>the</strong> alphasubunit<strong>of</strong> inhibin in granular cell tumors. Am J Clin Pathol119:259–26460. Fiocchi C (1998) Inflammatory bowel disease: etiology <strong>and</strong>pathogenesis. Gastroenterology 115:182–20561. Fornatora ML, Reich RF, Haber S, Solomon F, Freedman PD(2003) Oral melanoacanthoma: a report <strong>of</strong> 10 cases, review <strong>of</strong><strong>the</strong> literature, <strong>and</strong> immunohistochemical analysis for HMB-45 reactivity. Am J Dermatopathol 25:12–1562. Freysdottir J, Lau S, Fortune F (1999) Gammadelta T cellsin Behcet’s disease (BD) <strong>and</strong> recurrent aphthous stomatitis(RAS). Clin Exp Immunol 118:451–45763. Fukase S, Ohta N, Inamura K, Kimura Y, Aoyagi M, Koike Y(1994) Diagnostic specificity <strong>of</strong> anti-neutrophil cytoplasmicantibodies (ANCA) in otorhinolaryngological diseases. ActaOtolaryngol Suppl 511:204–20764. Goode RK, Crawford BE, Callihan MD, Neville BW (1983)Oral melanoacanthoma. Review <strong>of</strong> <strong>the</strong> literature <strong>and</strong> report<strong>of</strong> ten cases. Oral Surg Oral Med Oral Pathol 56:622–62865. Goulart RA, Mark EJ, Rosen S (1995) Tumefactions as an extravascularmanifestation <strong>of</strong> Wegener’s granulomatosis. AmJ Surg Pathol 19:145–15366. Grattan CE, Gentle TA, Basu MK (1992) Oral papillary plasmacytosisresembling c<strong>and</strong>idosis without demonstrable fungusin lesional tissue. Clin Exp Dermatol 17:112–11667. Greene FL, Page DL, Fleming ID, Fritz AG, Balch CM, HallerDG, Morrow M (2002) American Joint Committee on Cancer,Cancer Staging Manual. Springer, New York Berlin Heidelberg68. Greenstein RJ (2003) Is Crohn’s disease caused by a mycobacterium?Comparisons with leprosy, tuberculosis, <strong>and</strong>Johne’s disease. Lancet Infect Dis 3:507–51469. H<strong>and</strong>lers JP, Waterman J, Abrams AM, Melrose RJ (1985)Oral features <strong>of</strong> Wegener’s granulomatosis. Arch Otolaryngol111:267–27070. Harman KE, Seed PT, Gratian MJ, Bhogal BS, ChallacombeSJ, Black MM (2001) The severity <strong>of</strong> cutaneous <strong>and</strong> oral pemphigusis related to desmoglein 1 <strong>and</strong> 3 antibody levels. Br JDermatol 144:775–78071. Harrison JD, Maretin IC (1986) Oral vegetable granuloma:ultrastructural <strong>and</strong> histological study. J Oral Pathol 15:322-32672. Hedin CA, Pindborg JJ, Axell T (1993) Disappearance <strong>of</strong>smoker’s melanosis after reducing smoking. J Oral PatholMed 22:228–23073. Hegarty AM, Barrett AW, Scully C (2004) Pyostomatitis vegetans.Clin Exp Dermatol 29:1–774. Hickman RE, Eveson JW, Cawson RA (1988) Nevus uniuslateris <strong>and</strong> intraoral verrucous nevi. Oral Surg Oral MedOral Pathol 66:226–22975. Hicks MJ, Flaitz CM (2000) Oral mucosal melanoma: epidemiology<strong>and</strong> pathobiology. Oral Oncol 36:152–16976. H<strong>of</strong>fman GS (1994) “Wegener’s granulomatosis”: The pathtravelled since 1931. Medicine 73:325–32977. Honma T, Saito T, Fujioka Y (1985) Possible role <strong>of</strong> apoptoticcells <strong>of</strong> <strong>the</strong> oral epi<strong>the</strong>lium in <strong>the</strong> pathogenesis <strong>of</strong> aphthousulceration. Oral Surg Oral Med Oral Pathol 59:379–38778. Horn TD, Anhalt GJ (1992) Histologic features <strong>of</strong> paraneoplasticpemphigus. Arch Dermatol 128:1091–109579. Houston GD (1982) The giant cell fibroma. A review <strong>of</strong> 464cases. Oral Surg Oral Med Oral Pathol 53:582–58780. Hume G, Radford-Smith GL (2002) The pathogenesis <strong>of</strong>Crohn’s disease in <strong>the</strong> 21st century. <strong>Pathology</strong> 34:561–56781. Hume WJ (1975) Geographic stomatitis: a critical review. JDent 3:25–4382. Hume WJ, Smith CJ, Franklin CD (1980) Verruciform xanthoma.Br J Oral Surg 18:157–16183. Ide F, Mishima K, Saito I (2003) Juxtaoral organ <strong>of</strong> Chievitzpresenting clinically as a tumour. J Clin Pathol 56:789–79084. Itonaga I, Hussein I, Kudo O, Sabokbar A, Watt-Smith S, FergusonD, Athanasou NA (2003) Cellular mechanisms <strong>of</strong> osteoclastformation <strong>and</strong> lacunar resorption in giant cell granuloma<strong>of</strong> <strong>the</strong> jaw. J Oral Pathol Med 32:224–23185. Jabbour SA (2003) Cutaneous manifestations <strong>of</strong> endocrinedisorders: a guide for dermatologists. Am J Clin Dermatol4:315–331


100 J.W. Eveson386. James J, Patton DW, Lewis CJ et al. (1986) Oro-facial granulomatosis<strong>and</strong> clinical atopy. J Oral Med 41:29–3087. Jordan RC, Regezi JA (2003) Oral spindle cell neoplasms: areview <strong>of</strong> 307 cases. Oral Surg Oral Med Oral Pathol Oral RadiolEndod 95:717–72488. Jorizzo JL, Rogers RS III (1990) Behcet’s disease. J Am AcadDermatol 23:738–74189. Kanitakis J, Wang YZ, Roche P, Cozzani E et al. (1994) Immunohistopathologicalstudy <strong>of</strong> autoimmune pemphigus.Lack <strong>of</strong> strictly specific histological <strong>and</strong> indirect immun<strong>of</strong>luorescencecriteria for paraneoplastic pemphigus. Dermatology188:282–28590. Karjalainen TK, Tomich CE (1989) A histopathologic study<strong>of</strong> oral mucosal lupus ery<strong>the</strong>matosus. Oral Surg Oral MedOral Pathol 67:547–55491. Kennedy TL (1989) Cystic hygroma-lymphangioma: a rare<strong>and</strong> still unclear entity. Laryngoscope 99:1–1092. Kimyai-Asadi A, Jih MH (2001) Paraneoplastic pemphigus.Int J Dermatol 40:367–37293. Kitagawa S, Townsend BL, Hebert AA (1995) Peutz-Jegherssyndrome. Dermatol Clin 13:127–13394. Kramer IR, El-Labban N, Lee KW (1978) The clinical features<strong>and</strong> risk <strong>of</strong> malignant transformation in sublingualkeratosis. Br Dent J 144:171–18095. Lamey PJ, Carmichael F, Scully C (1985) Oral pigmentation,Addison’s disease <strong>and</strong> <strong>the</strong> results <strong>of</strong> screening for adrenocorticalinsufficiency. Br Dent J 158:297–29896. Lange J de, Rosenberg AJ, Akker HP van den, Koole R, WirdsJJ, Berg H van den (1999) Treatment <strong>of</strong> central giant cellgranuloma <strong>of</strong> <strong>the</strong> jaw with calcitonin. Int J Oral Maxill<strong>of</strong>acSurg 28:372–37697. Lehner T (1969) <strong>Pathology</strong> <strong>of</strong> recurrent oral ulceration <strong>and</strong>oral ulceration in Behcet’s syndrome: light, electron <strong>and</strong> fluorescencemicroscopy. J Pathol 97:481–49498. Lenane P, Powell FC (2000) Oral pigmentation. J Eur AcadDermatol Venereol 14:448–46599. Levy M, Shear NH (1991) Mycoplasma pneumoniae infections<strong>and</strong> Stevens-Johnson syndrome. Report <strong>of</strong> eight cases<strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Clin Pediatr (Phila) 30:42–49100. Liu AY, Valenzuela R, Helm TN, Camisa C et al. (1993) Indirectimmun<strong>of</strong>luorescence on rat bladder transitional epi<strong>the</strong>lium:a test with high specificity for paraneoplastic pemphigus.J Am Acad Dermatol 28:696–699101. Liu B, Yu SF, Li TJ (2003) Multinucleated giant cells in variousforms <strong>of</strong> giant cell containing lesions <strong>of</strong> <strong>the</strong> jaws expressfeatures <strong>of</strong> osteoclasts. J Oral Pathol Med 32:367–375102. Lopez R, Fern<strong>and</strong>ez O, Jara G, Baelum V (2002) Epidemiology<strong>of</strong> necrotizing ulcerative gingival lesions in adolescents. JPeriodontal Res 37:439–444103. Lutman GB (1974) Epi<strong>the</strong>lial nests in intraoral sensory nerveendings simulating perineural invasion in patients with oralcarcinoma. Am J Clin Pathol 61:275–284104. McCartan BE, McCreary CE (1997) Oral lichenoid drugeruptions. Oral Dis 3:58–63105. McCartan BE, McCreary CE, Healy CM (2003) Studies <strong>of</strong>drug-induced lichenoid reactions: criteria for case selection.Oral Dis 9:163–164106. McGregor AD, MacDonald DG (1988) Routes <strong>of</strong> entry <strong>of</strong>squamous cell carcinoma into <strong>the</strong> m<strong>and</strong>ible. <strong>Head</strong> <strong>Neck</strong>10:294–301107. Magnusson BC, Rasmusson LG (1995) The giant cell fibroma.A review <strong>of</strong> 103 cases with immunohistochemical findings.Acta Odontol Sc<strong>and</strong> 53:293–296108. Maiorano E, Favia G, Napoli A, Resta L, Ricco R, Viale G,Altini M (2000) Cellular heterogeneity <strong>of</strong> granular cell tumours:a clue to <strong>the</strong>ir nature? J Oral Pathol Med 29:284–290109. Main DM, Chamberlain MA (1992) Clinical differentiation<strong>of</strong> oral ulceration in Behcet’s disease. Br J Rheumatol 31:767–770110. Manabe M, Lim HW, Winzer M, Loomis CA (1999) Architecturalorganization <strong>of</strong> filiform papillae in normal <strong>and</strong> blackhairy tongue epi<strong>the</strong>lium: dissection <strong>of</strong> differentiation pathwaysin a complex human epi<strong>the</strong>lium according to <strong>the</strong>ir patterns<strong>of</strong> keratin expression. Arch Dermatol 135:177–181111. Manch<strong>and</strong>a Y, Tejasvi T, H<strong>and</strong>a R, Ramam M (2003) Strawberrygingiva: a distinctive sign in Wegener’s granulomatosis.J Am Acad Dermatol 49:335–337112. Mani NJ (1985) Tuberculosis initially diagnosed by asymptomaticoral lesions. Report <strong>of</strong> three cases. J Oral Med 40:39–42113. Manus DA, Sherbert D, Jackson IT (1995) Management considerationsfor <strong>the</strong> granuloma <strong>of</strong> pregnancy. Plast ReconstrSurg 95:1045–1050114. Marks R, Radden BG (1981) Geographic tongue: a clinicopathologicalreview. Australas J Dermatol 22:75–79115. Mashberg A, Samit A (1995) Early diagnosis <strong>of</strong> symptomaticoral <strong>and</strong> orophayngeal squamous cancers. CA Cancer J Clin45:328–351116. Meador R, Ehrlich G, Von Feldt JM (2002) Behcet’s disease:immunopathologic <strong>and</strong> <strong>the</strong>rapeutic aspects. Curr RheumatolRep 4:47–54117. Merchant HW, Hayes LE, Ellison LT (1976) S<strong>of</strong>t-palate pigmentationin lung disease, including cancer. Oral Surg OralMed Oral Pathol 41:726–733118. Mignogna MD, Lo Muzio L, Ruoppo E, Errico M, Amato M,Satriano RA (1999) Oral manifestations <strong>of</strong> idiopathic lenticularmucocutaneous pigmentation (Laugier-Hunziker syndrome):a clinical, histopathological <strong>and</strong> ultrastructural review<strong>of</strong> 12 cases. Oral Dis 1999 5:80–86119. Miles AEW (1958) Sebaceous gl<strong>and</strong>s in <strong>the</strong> lip <strong>and</strong> cheek mucosa<strong>of</strong> man. Br Dent J 105:235–248120. Mooney EE, Walker J, Hourihane DO (1995) Relation <strong>of</strong>granulomas to lymphatic vessels in Crohn’s disease. J ClinPathol 48:335–338121. Mostafa KA, Takata T, Ogawa I, Ijuhin N, Nikai H (1993)Verruciform xanthoma <strong>of</strong> <strong>the</strong> oral mucosa: a clinicopathologicalstudy with immunohistochemical findings relatingto pathogenesis. Virchows Arch A Pathol Anat Histopathol423:243–248122. Murray A, Gardiner DS, McGuiness RJ (1998) Primary mycobacterialinfection <strong>of</strong> <strong>the</strong> uvula. J Laryngol Otol 112:1183–1185123. N<strong>and</strong>apalan V, Rol<strong>and</strong> NJ, Helliwell TR, Williams EM, HamiltonJW, Jones AS (1998) Mucosal melanoma <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck. Clin Otolaryngol 23:107–116124. Napier SS, Allen JA, Irwin CR, McCluskey DR (1993) Strawberrygums: a clinicopathological manifestation diagnostic<strong>of</strong> Wegener’s granulomatosis? J Clin Pathol 46:709–712125. Neville BW, Wea<strong>the</strong>rs DR (1980) Verruciform xanthoma.Oral Surg Oral Med Oral Pathol 49:429–434126. Nicolas ME, Krause PK, Gibson LE, Murray JA (2003) Dermatitisherpetiformis. Int J Dermatol 42:588–600127. Nowparast B, Howell FV, Rick GM (1981) Verruciform xanthoma.A clinicopathologic review <strong>and</strong> report <strong>of</strong> fifty-fourcases. Oral Surg Oral Med Oral Pathol 51:619–625128. Nozicka Z (1985) Endovasal granulomatous lymphangiitisas a pathogenetic factor in cheilitis granulomatosa. J OralPathol 14:363–365129. Ogus HD, Bennett MH (1978) Carcinoma <strong>of</strong> <strong>the</strong> dorsum <strong>of</strong><strong>the</strong> tongue: a rarity or misdiagnosis. Br J Oral Surg 16:115–124130. Obermayer-Straub P, Manns MP (1998) Autoimmune polygl<strong>and</strong>ularsyndromes. Baillieres Clin Gastroenterol 12:293–315131. O’Keefe E, Braverman IM, Cohen I (1973) Annulus migrans.Identical lesions in pustular psoriasis, Reiter’s syndrome, <strong>and</strong>geographic tongue. Arch Dermatol 107:240–244132. Pantanowitz L, Balogh K (2003) Significance <strong>of</strong> <strong>the</strong> juxtaoralorgan (<strong>of</strong> Chievitz). <strong>Head</strong> <strong>Neck</strong> 25:400–405133. Park YW, Kim SM, Min BG, Park IW, Lee SK (2002) Lymphangiomainvolving <strong>the</strong> m<strong>and</strong>ible: immunohistochemical expressionsfor <strong>the</strong> lymphatic proliferation. J Oral Pathol Med31:280–283134. Parkin DM, Whelan SL, Ferlay J et al. (2003) Cancer incidencein five continents, vol. VIII. IARC Press, Lyon135. Philipsen HP, Reichart PA, Takata T, Ogawa I (2003) Verruciformxanthoma – biological pr<strong>of</strong>ile <strong>of</strong> 282 oral lesionsbased on a literature survey with nine new cases from Japan.Oral Oncol 39:325–336


Oral Cavity Chapter 3 101136. Pindborg JJ, Reibel J, Roed-Peterson B, Mehta FS (1980) Tobacco-inducedchanges in oral leukoplakic epi<strong>the</strong>lium. Cancer45:2330–2336137. Pinto GM, Lamarao P, Vale T (1992) Captopril-induced pemphigusvegetans with Charcot-Leyden crystals. Am AcadDermatol 27:281–284138. Plauth M, Jenss H, Meyle J (1991) Oral manifestations <strong>of</strong>Crohn’s disease. An analysis <strong>of</strong> 79 cases. J Clin Gastroenterol13:29–37139. Pogrel MA (1979) Sublingual keratosis <strong>and</strong> malignant transformation.J Oral Pathol 8:176–178140. Pola JG, de la Cruz, Bustillo F, Gallas M, Leston JS (2003)Pulse granuloma in <strong>the</strong> wall <strong>of</strong> an inflammatory radicularcyst. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 129:441–442141. Popa ER, Tervaert JW (2003) The relation between Staphylococcusaureus <strong>and</strong> Wegener’s granulomatosis: currentknowledge <strong>and</strong> future directions. Intern Med 42:771–780142. Porter SR, Hegarty A, Kaliakatsou F, Hodgson TA, ScullyC (2000) Recurrent aphthous stomatitis. Clin Dermatol18:569–578143. Porter SR, Scully C, Pedersen A (1998) Recurrent aphthousstomatitis. Crit Rev Oral Biol Med 9:306–321144. Prasad ML, Jungbluth AA, Iversen K, Huvos AG, Busam KJ(2001) Expression <strong>of</strong> melanocytic differentiation markers inmalignant melanomas <strong>of</strong> <strong>the</strong> oral <strong>and</strong> sinonasal mucosa. AmJ Surg Pathol 25:782–787145. Prasad ML, Patel S, Hoshaw-Woodard S et al. (2002) Prognosticfactors for malignant melanoma <strong>of</strong> <strong>the</strong> squamous mucosa<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Am J Surg Pathol 26:883–928146. Pryce DW, King CM (1990) Or<strong>of</strong>acial granulomatosis associatedwith delayed hypersensitivity to cobalt. Clin Exp Dermatol15:384–386147. Pujol RM, Matias-Guiu X, Miralles J, Colomer A, de MoragasJM (1997) Multiple idiopathic mucosal neuromas: a minorform <strong>of</strong> multiple endocrine neoplasia type 2B or a newentity? J Am Acad Dermatol 337:349–352148. Quigley LF, Cobb CM, Hunt EE, Williams P, Schoenfeld S(1964) Reverse smoking <strong>and</strong> its oral consequences in Caribbean<strong>and</strong> South American peoples. J Am Dent Assoc 69:426–442149. Reichart PA, Langford A, Gelderblom HR et al. (1989) Oralhairy leukoplakia: observations in 95 cases <strong>and</strong> review <strong>of</strong> <strong>the</strong>literature. J Oral Pathol Med 18:410–415150. Reichart PA, Samaranayake LP, Philipsen HP (2000) <strong>Pathology</strong><strong>and</strong> clinical correlates in oral c<strong>and</strong>idiasis <strong>and</strong> its variants:a review. Oral Dis 6:85–91151. Reichart PA, Schmidt-Westhausen A, Samaranayake LP,Philipsen HP (1994) C<strong>and</strong>ida-associated palatal papillaryhyperplasia in HIV infection. J Oral Pathol Med 23:403–405152. Rennie JS, Reade PC, Hay KD, Scully C (1985) Recurrentaphthous stomatitis. Br Dent J 159:361–367153. Reynolds RM, Browning GG, Nawroz I, Campbell IW (2003)Von Recklinghausen’s neur<strong>of</strong>ibromatosis: neur<strong>of</strong>ibromatosistype 1. Lancet 361:1552–1554154. Rogers RS III (1997) Recurrent aphthous stomatitis: clinicalcharacteristics <strong>and</strong> associated systemic disorders. Semin CutanMed Surg 16:278–283155. Sah SP, Raj GA, Bahadur T (1999) Chronic ulceration <strong>of</strong> <strong>the</strong>tongue <strong>and</strong> laryngitis: first clinical sign <strong>of</strong> asymptomaticpulmonary tuberculosis. J Infect 39:163–164156. Schiodt M (1984) Oral discoid lupus ery<strong>the</strong>matosus. III. Ahistopathologic study <strong>of</strong> sixty-six patients. Oral Surg OralMed Oral Pathol 57:281–293157. Schiodt M, Halberg P, Hentzer B (1978) A clinical study <strong>of</strong>32 patients with oral discoid lupus ery<strong>the</strong>matosus. Int J OralSurg 7:85–94158. Schwartz DL (1965) Stomatitis nicotina <strong>of</strong> <strong>the</strong> palate; report<strong>of</strong> two cases. Oral Surg Oral Med Oral Pathol 20:306–315159. Sciubba JJ, Said-Al-Naief N (2003) Or<strong>of</strong>acial granulomatosis:presentation, pathology <strong>and</strong> management <strong>of</strong> 13 cases. J OralPathol Med 32:576–585160. Scott JE, Ahmed AR (1998) The blistering diseases. Med ClinNorth Am 82:1239–1283161. Scott LA, Stone MS (2003) Viral exan<strong>the</strong>ms. Dermatol OnlineJ 9:4162. Scully C, Beyli M, Ferreiro MC et al. (1998) Update on orallichen planus: etiopathogenesis <strong>and</strong> management. Crit RevOral Biol Med 9:86–122163. Scully C, Challacombe SJ (2002) Pemphigus vulgaris: updateon etiopathogenesis, oral manifestations, <strong>and</strong> management.Crit Rev Oral Biol Med 13:397–408164. Sedghizadeh PP, Shuler CF, Allen CM, Beck FM, Kalmar JR(2002) Celiac disease <strong>and</strong> recurrent aphthous stomatitis: areport <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Oral Surg Oral Med OralPathol Oral Radiol Endod 94:474–478165. Sela MN (2001) Role <strong>of</strong> Treponema denticola in periodontaldiseases. Crit Rev Oral Biol Med 12:399–413166. Shafer WG (1971) Verruciform xanthoma. Oral Surg OralMed Oral Pathol 31:784–789167. Sobin LH, Wittekind C (2002) TNM: classification <strong>of</strong> malignanttumours, 6th edn. Wiley, New York168. Southam JC, Felix DH, Wray D, Cubie HA (1991) Hairy leukoplakia– a histological study. Histopathology 19:63–67169. Sweatman MC, Tasker R, Warner JO et al. (1986) Oro-facialgranulomatosis. response to elemental diet <strong>and</strong> provocationby food additives. Clin Allergy 16:331–338170. Sund S, Bang G (1990) Intramuscular hemangioma in <strong>the</strong>oral region: report <strong>of</strong> three cases. Oral Surg Oral Med OralPathol 70:765–768171. Talacko AA, Radden BG (1988) Oral pulse granuloma: clinical<strong>and</strong> histopathological features. A review <strong>of</strong> 62 cases. Int JOral Maxill<strong>of</strong>ac Surg 17:343–346172. Thomas JA, Felix DH, Wray D et al. (1991) Epstein-Barr virusgene expression <strong>and</strong> epi<strong>the</strong>lial cell differentiation in oralhairy leukoplakia. Am J Pathol 139:1369–1380173. Thompson DE (1994) The role <strong>of</strong> mycobacteria in Crohn’sdisease. J Med Microbiol 41:74–94174. Tucker KM, Heget HS (1976) The incidence <strong>of</strong> inflammatorypapillary hyperplasia. J Am Dent Assoc 93:610–613175. Tsuboi H, Fujimura T, Katsuoka K (2001) Angiolymphoidhyperplasia with eosinophilia in <strong>the</strong> oral mucosa. Br J Dermatol145:365–366176. Umeda M, Komatsubara H, Shibuya Y, Yokoo S, Komori T(2002) Premalignant melanocytic dysplasia <strong>and</strong> malignantmelanoma <strong>of</strong> <strong>the</strong> oral mucosa. Oral Oncol 38:714–722177. Verpilleux MP, Bastuji-Garin S, Revuz J (1999) Comparativeanalysis <strong>of</strong> severe aphthosis <strong>and</strong> Behcet’s disease: 104 cases.Dermatology 198:247–251178. Wakefield AJ, Sankey EA, Dhillon AP et al. (1991) Granulomatousvasculitis in Crohn’s disease. Gastroenterology100:1279–1287179. Wal N van der, Kwast WA van der, Dijk E van, Waal I van der(1988) Geographic stomatitis <strong>and</strong> psoriasis. Int J Oral Maxill<strong>of</strong>acSurg 17:106–109180. Wal N van der, Waal I van der (1986) C<strong>and</strong>ida albicans inmedian rhomboid glossitis. A postmortem study. Int J OralMaxill<strong>of</strong>ac Surg 15:322–325181. Weiss SW, Golblum JR (2001) Benign tumors <strong>and</strong> tumor-likelesions <strong>of</strong> blood vessels. In: Enzinger <strong>and</strong> Weiss’s s<strong>of</strong>t tissuetumors, 4th edn. Mosby, St. Louis, pp 840–843182. Weitzul S, Duvic M (1997) HIV-related psoriasis <strong>and</strong> Reiter’ssyndrome. Semin Cutan Med Surg 16:213–218183. Whitley RJ (2002) Herpes simplex virus infection. Semin PediatrInfect Dis 13:6–11184. Wiesenfeld D, Ferguson MM, Mitchell DN et al. (1985) Or<strong>of</strong>acialgranulomatosis – a clinical <strong>and</strong> pathological analysis.Q J Med 54:101–113185. Williams AJ, Wray D, Ferguson A (1991) The clinical entity<strong>of</strong> or<strong>of</strong>acial Crohn’s disease. Q J Med 79:451–458186. Williams DM (1990) Vesiculo-bullous mucocutaneous disease:benign mucous membrane <strong>and</strong> bullous pemphigoid. JOral Pathol Med 19:16–23187. Woo TY, Solomon AR, Fairley JA (1985) Pemphigus vegetanslimited to <strong>the</strong> lips <strong>and</strong> oral mucosa. Arch Dermatol 121:271–272188. Wright BA, Fenwick F (1981) C<strong>and</strong>idiasis <strong>and</strong> atrophictongue lesions. Oral Surg Oral Med Oral Pathol 51:55–61


102 J.W. Eveson3189. Wright BA, Jackson D (1980) Neural tumors <strong>of</strong> <strong>the</strong> oral cavity.A review <strong>of</strong> <strong>the</strong> spectrum <strong>of</strong> benign <strong>and</strong> malignant oraltumors <strong>of</strong> <strong>the</strong> oral cavity <strong>and</strong> jaws. Oral Surg Oral Med OralPathol 49:509–522190. Yamamoto O, Yoshinaga K, Asahi M, Murata I (1999) A Laugier-Hunzikersyndrome associated with esophageal melanocytosis.Dermatology 199:162–164191. Yeh SW, Ahmed B, Sami N, Razzaque Ahmed A (2003) Blisteringdisorders: diagnosis <strong>and</strong> treatment. Dermatol Ther16:214–223192. Zimmer WM, Rogers RS 3rd, Reeve CM, Sheridan PJ (1992)Or<strong>of</strong>acial manifestations <strong>of</strong> Melkersson-Rosenthal syndrome.A study <strong>of</strong> 42 patients <strong>and</strong> review <strong>of</strong> 220 cases from<strong>the</strong> literature. Oral Surg Oral Med Oral Pathol 74:610–619193. Zunt SL (2003) Recurrent aphthous stomatitis. DermatolClin 21:33–39194. Zunt SL, Tomich CE (1989) Ery<strong>the</strong>ma migrans – a psoriasiformlesion <strong>of</strong> <strong>the</strong> oral mucosa. J Dermatol Surg Oncol15:1067–1070


Chapter 4Maxill<strong>of</strong>acial Skeleton<strong>and</strong> TeethP.J. Slootweg4Contents4.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . 1044.1.1 Embryology . . . . . . . . . . . . . . . . . . . . . . . 1044.1.2 Tooth Development . . . . . . . . . . . . . . . . . . . 1044.2 Inflammatory Diseases<strong>of</strong> <strong>the</strong> Maxill<strong>of</strong>acial Bones . . . . . . . . . . . . . . . 1044.3 Cysts <strong>of</strong> <strong>the</strong> Jaws . . . . . . . . . . . . . . . . . . . . . 1054.3.1 Odontogenic Cysts – Inflammatory . . . . . . . . . 1054.3.1.1 Radicular Cyst . . . . . . . . . . . . . . . . . . . . . . 1054.3.1.2 Paradental Cyst . . . . . . . . . . . . . . . . . . . . . 1064.3.2 Odontogenic Cysts – Developmental . . . . . . . . . 1064.3.2.1 Dentigerous Cyst . . . . . . . . . . . . . . . . . . . . 1064.3.2.2 Lateral Periodontal Cyst . . . . . . . . . . . . . . . . 1074.3.2.3 Gl<strong>and</strong>ular Odontogenic Cyst . . . . . . . . . . . . . 1074.3.2.4 Odontogenic Keratocyst . . . . . . . . . . . . . . . . 1074.3.2.5 Gingival Cyst . . . . . . . . . . . . . . . . . . . . . . . 1084.3.3 Non-Odontogenic Cysts . . . . . . . . . . . . . . . . 1094.3.3.1 Nasopalatine Duct Cyst . . . . . . . . . . . . . . . . 1094.3.3.2 Nasolabial Cyst . . . . . . . . . . . . . . . . . . . . . 1094.3.3.3 Surgical Ciliated Cyst . . . . . . . . . . . . . . . . . . 1094.3.4 Pseudocysts . . . . . . . . . . . . . . . . . . . . . . . 1094.3.4.1 Solitary Bone Cyst . . . . . . . . . . . . . . . . . . . . 1094.3.4.2 Focal Bone Marrow Defect . . . . . . . . . . . . . . . 1094.4 Odontogenic Tumours . . . . . . . . . . . . . . . . . 1094.4.1 Odontogenic Tumours – Epi<strong>the</strong>lial . . . . . . . . . . 1104.4.1.1 Ameloblastoma . . . . . . . . . . . . . . . . . . . . . 1104.4.1.2 Calcifying Epi<strong>the</strong>lial Odontogenic Tumour . . . . . 1124.4.1.3 Adenomatoid Odontogenic Tumour . . . . . . . . . 1124.4.1.4 Squamous Odontogenic Tumour . . . . . . . . . . . 1134.4.2 Odontogenic Tumours – Mesenchymal . . . . . . . 1144.4.2.1 Odontogenic Myxoma . . . . . . . . . . . . . . . . . 1144.4.2.2 Odontogenic Fibroma . . . . . . . . . . . . . . . . . . 1154.4.2.3 Cementoblastoma . . . . . . . . . . . . . . . . . . . . 1164.4.3 Odontogenic Tumours –Mixed Epi<strong>the</strong>lial <strong>and</strong> Mesenchymal . . . . . . . . . 1174.4.3.1 Ameloblastic Fibroma . . . . . . . . . . . . . . . . . 1174.4.3.2 Ameloblastic Fibro-Odontoma . . . . . . . . . . . . 1174.4.3.3 Odontoma – Complex Type . . . . . . . . . . . . . . 1184.4.3.4 Odontoma – Compound Type . . . . . . . . . . . . . 1184.4.3.5 Odonto-Ameloblastoma . . . . . . . . . . . . . . . . 1184.4.3.6 Calcifying Odontogenic Cyst . . . . . . . . . . . . . 1194.4.4 Odontogenic Tumours – Malignant . . . . . . . . . 1194.4.4.1 Malignant Ameloblastoma . . . . . . . . . . . . . . . 1194.4.4.2 Ameloblastic Carcinoma . . . . . . . . . . . . . . . . 1194.4.4.3 Primary Intraosseous Carcinoma . . . . . . . . . . . 1194.4.4.4 Clear Cell Odontogenic Carcinoma . . . . . . . . . . 1204.4.4.5 Malignant Epi<strong>the</strong>lial OdontogenicGhost Cell Tumour . . . . . . . . . . . . . . . . . . . 1204.4.4.6 Odontogenic Sarcoma . . . . . . . . . . . . . . . . . 1204.5 Fibro-Osseous Lesions . . . . . . . . . . . . . . . . . 1214.5.1 Fibrous Dysplasia . . . . . . . . . . . . . . . . . . . . 1214.5.2 Ossifying Fibroma . . . . . . . . . . . . . . . . . . . . 1214.5.3 Osseous Dysplasia . . . . . . . . . . . . . . . . . . . . 1234.6 Giant Cell Lesions . . . . . . . . . . . . . . . . . . . . 1244.6.1 Central Giant Cell Granuloma . . . . . . . . . . . . 1244.6.2 Cherubism . . . . . . . . . . . . . . . . . . . . . . . . 1244.7 Neoplastic Lesions <strong>of</strong> <strong>the</strong> Maxill<strong>of</strong>acial Bones,Non-Odontogenic . . . . . . . . . . . . . . . . . . . . 1254.7.1 Osteoma . . . . . . . . . . . . . . . . . . . . . . . . . 1254.7.2 Chordoma . . . . . . . . . . . . . . . . . . . . . . . . 1254.7.3 Melanotic Neuroectodermal Tumour<strong>of</strong> Infancy . . . . . . . . . . . . . . . . . . . . . . . . . 126References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 126


104 P.J. Slootweg4.1 Introduction4.1.1 Embryology4The maxill<strong>of</strong>acial skeleton is partly derived from migratedcranial neural crest cells. These cells give riseto most connective tissues in <strong>the</strong> crani<strong>of</strong>acial regionincluding <strong>the</strong> bones <strong>of</strong> <strong>the</strong> calvarium, face <strong>and</strong> jaws.So, <strong>the</strong> diseases to be discussed in this chapter occurin bones that are formed by mesenchymal cells withan ectodermal/neuroectodermal ancestry – that iswhy <strong>the</strong>y are also known as ectomesenchyme – <strong>and</strong>are <strong>the</strong>refore different from bone <strong>and</strong> cartilage thathave a mesodermal origin elsewhere in <strong>the</strong> body [145,171].4.1.2 Tooth DevelopmentTeeth develop from epi<strong>the</strong>lial cells from <strong>the</strong> mucosal lining<strong>of</strong> <strong>the</strong> oral cavity <strong>and</strong> cranial neural crest-derived ectomesenchymalcells. Under <strong>the</strong> influence <strong>of</strong> reciprocalinductive events, <strong>the</strong>se cells develop into enamel-formingameloblasts <strong>and</strong> dentin-producing odontoblasts(Fig. 4.1) [145, 171].While ameloblasts <strong>and</strong> odontoblasts are depositingenamel <strong>and</strong> dentin, <strong>the</strong> epi<strong>the</strong>lium proliferates downwards,thus creating a tube that maps out <strong>the</strong> form <strong>and</strong>size <strong>of</strong> <strong>the</strong> root <strong>of</strong> <strong>the</strong> teeth. This epi<strong>the</strong>lial cuff is knownas <strong>the</strong> sheath <strong>of</strong> Hertwig. Its remnants form a permanentcomponent <strong>of</strong> <strong>the</strong> periodontal ligament; <strong>the</strong>y are knownas rests <strong>of</strong> Malassez <strong>and</strong> are <strong>the</strong> source <strong>of</strong> some cysticjaw lesions (Fig. 4.2). O<strong>the</strong>r epi<strong>the</strong>lial reminiscences <strong>of</strong>tooth development lie more superficially in <strong>the</strong> jaw tissues.These are <strong>the</strong> epi<strong>the</strong>lial rests <strong>of</strong> Serres, which have<strong>the</strong>ir origin in <strong>the</strong> dental lamina.4.2 Inflammatory Diseases<strong>of</strong> <strong>the</strong> Maxill<strong>of</strong>acial BonesThese include osteomyelitis, which mainly involves <strong>the</strong>m<strong>and</strong>ible. It may occur through extension <strong>of</strong> infection<strong>of</strong> <strong>the</strong> dental pulp or as a complication after tooth extraction.When seen after irradiation, e.g. for head <strong>and</strong>neck cancer, it is called osteoradionecrosis.There are five types: acute suppurative osteomyelitis,chronic suppurative osteomyelitis, chronic focal sclerosingosteomyelitis, chronic diffuse sclerosing osteomyelitis,<strong>and</strong> proliferative periostitis. Acute suppurative osteomyelitisshows bone marrow cavities infiltrated withneutrophils. The bony trabeculae are necrotic. Usually,this form <strong>of</strong> osteomyelitis evolves into chronic suppurativeosteomyelitis, which also may arise de novo. Besidesbone sequesters surrounded by numerous neutrophilicFig. 4.1. Overview <strong>of</strong> normal tooth germ lying in its bony crypt.Odontogenic epi<strong>the</strong>lium covers <strong>the</strong> enamel space from which <strong>the</strong>enamel is dissolved due to decalcification. An epi<strong>the</strong>lial str<strong>and</strong>as remnant <strong>of</strong> <strong>the</strong> dental lamina can be seen in <strong>the</strong> fibrous tissuebridging <strong>the</strong> gap in <strong>the</strong> bony cryptFig. 4.2. The periodontal ligament connects <strong>the</strong> root surface(right side) with <strong>the</strong> bony socket (left side). An epi<strong>the</strong>lial rest <strong>of</strong>Malassez is clearly visiblegranulocytes granulation tissue is also present. Sinusesare formed partly lined by squamous epi<strong>the</strong>lium from<strong>the</strong> oral mucosa. In less severe cases, fibrosis <strong>and</strong> development<strong>of</strong> a chronic inflammatory infiltrate may alsobe seen. Unless <strong>the</strong> sequestra are removed, <strong>the</strong> diseasewill not heal.When <strong>the</strong> inflammation is mild, <strong>the</strong> jaw bone respondsby bone formation. This form <strong>of</strong> osteomyelitis isknown as chronic sclerosing, which may be focal as wellas diffuse. Dense sclerotic bone masses are seen toge<strong>the</strong>rwith a bone marrow exhibiting oedema <strong>and</strong> small foci<strong>of</strong> lymphocytes <strong>and</strong> plasma cells. Both focal as well asdiffuse chronic sclerosing osteomyelitis must be distinguishedfrom o<strong>the</strong>r bone lesions, especially <strong>the</strong> fibro-osseousones (see Sect. 4.5).


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 105Table 4.1. Cysts <strong>of</strong> <strong>the</strong> jaws [73]Odontogenic cysts – inflammatoryOdontogenic cysts – developmentalNon-odontogenic cystsPseudocystsRadicular cystResidual cystParadental cystDentigerous cystLateral periodontal cystBotryoid odontogenic cystGl<strong>and</strong>ular odontogenic cystOdontogenic keratocyst (keratocystic odontogenic tumour [181])Gingival cystNasopalatine duct cystNasolabial cystSurgical ciliated cystSolitary bone cystFocal bone marrow defectWhen <strong>the</strong> inflammation mainly involves <strong>the</strong> periosteum,<strong>the</strong> disease is called proliferative periostitis, orcalled periostitis ossificans. Histologically, bony trabeculaethat lie in a linear parallel pattern are seen. The interveningstroma is composed <strong>of</strong> fibrous connective tissuesparsely infiltrated with lymphocytes <strong>and</strong> plasmacells.4.3 Cysts <strong>of</strong> <strong>the</strong> JawsCysts <strong>of</strong> <strong>the</strong> jaws are classified into several categories dependingon histogenesis <strong>and</strong> aetiology. Those that arisefrom odontogenic epi<strong>the</strong>lium are called odontogenic,those that have <strong>the</strong>ir source in o<strong>the</strong>r epi<strong>the</strong>lial structuresare known as non-odontogenic. Among <strong>the</strong> odontogeniccysts, developmental <strong>and</strong> inflammatory typescan be distinguished [73]. By definition, cysts are linedby epi<strong>the</strong>lium. There are, however, also cavities in <strong>the</strong>jaw lacking such an epi<strong>the</strong>lial investment that are alsodiscussed under this heading. The various entities arelisted in Table 4.1.4.3.1 Odontogenic Cysts –Inflammatory4.3.1.1 Radicular CystFig. 4.3. Schematic drawing showing a bisected m<strong>and</strong>ible. Left toright: examples <strong>of</strong> a dentigerous cyst, a lateral periodontal cyst <strong>and</strong>a radicular cyst (drawing by John de Groot)Radicular cysts are located at <strong>the</strong> root tips <strong>of</strong> <strong>the</strong> teeth(Fig. 4.3). They arise from <strong>the</strong> epi<strong>the</strong>lial rests <strong>of</strong> Malassez<strong>and</strong> are <strong>the</strong> cysts most frequently seen [34, 146].They are lined by non-keratinising squamous epi<strong>the</strong>lium.This epi<strong>the</strong>lial lining may be thin <strong>and</strong> atrophicor show elongated rete processes. In many cysts,cholesterol clefts with adjacent giant cells occur. Within<strong>the</strong> cyst epi<strong>the</strong>lium, hyaline bodies <strong>of</strong> various size<strong>and</strong> shape may be present (Fig. 4.4). The specific nature<strong>of</strong> <strong>the</strong>se so-called Rushton bodies is unclear [95]. Occasionally,<strong>the</strong> lining squamous cells are admixed withmucous cells or ciliated cells (Fig. 4.5). Sometimes, <strong>the</strong>histologic pattern <strong>of</strong> <strong>the</strong> radicular cyst is complicatedby extensive intramural proliferation <strong>of</strong> squamous epi<strong>the</strong>lialnests <strong>of</strong> varying size, thus mimicking a squamousodontogenic tumour (see Sect. 4.4.1.4) [187]. Thesame histology may be shown by o<strong>the</strong>r jaw cysts, in particularwhen <strong>the</strong>re are extensive inflammatory changes.When a radicular cyst is retained in <strong>the</strong> jaws afterremoval <strong>of</strong> <strong>the</strong> associated tooth, <strong>the</strong> lesion is called aresidual cyst .When complicated by inflammation, radicular cystsmay cause pain <strong>and</strong> swelling. However, <strong>the</strong>y may also beasymptomatic <strong>and</strong> fortuitously detected by radiographicexamination <strong>of</strong> <strong>the</strong> dentition.


106 P.J. Slootweg4Fig. 4.4. Epi<strong>the</strong>lial lining <strong>of</strong> a radicular cyst containing manyRushton bodiesFig. 4.5. Cyst lining composed <strong>of</strong> squamous as well as mucous epi<strong>the</strong>lium.This can be found in radicular cysts as well as in dentigerousor residual cysts4.3.1.2 Paradental CystThe paradental cyst is located on <strong>the</strong> lateral side <strong>of</strong> <strong>the</strong>tooth at <strong>the</strong> border between <strong>the</strong> enamel <strong>and</strong> root cementum.This cyst is secondary to an inflammatory processin <strong>the</strong> adjacent periodontal tissues that induces proliferation<strong>of</strong> neighbouring odontogenic epi<strong>the</strong>lial rests, similarto <strong>the</strong> pathogenesis <strong>of</strong> <strong>the</strong> radicular cyst [88]. Histologically,it resembles <strong>the</strong> o<strong>the</strong>r inflammatory odontogeniccysts, <strong>the</strong> distinction being made by <strong>the</strong> specificclinical presentation. It is a rare lesion [128]. Treatmentconsists <strong>of</strong> excision with or without concomitant extraction<strong>of</strong> <strong>the</strong> involved tooth [45].4.3.2 Odontogenic Cysts –Developmental4.3.2.1 Dentigerous CystA dentigerous cyst surrounds <strong>the</strong> crown <strong>of</strong> an uneruptedtooth, mostly <strong>the</strong> maxillary canine or <strong>the</strong> m<strong>and</strong>ibularthird molar tooth (Fig. 4.3). They are quite common.The cyst wall has a thin epi<strong>the</strong>lial lining that maybe only two to three cells thick. In case <strong>of</strong> inflammation,<strong>the</strong> epi<strong>the</strong>lium becomes thicker <strong>and</strong> will showfeatures similar to <strong>the</strong> lining <strong>of</strong> a radicular cyst. Also,mucous-producing cells as well as ciliated cells maybe observed (Fig. 4.6). The connective tissue component<strong>of</strong> <strong>the</strong> cyst wall may be fibrous or fibromyxomatous.The cyst wall may also contain varying amounts<strong>of</strong> epi<strong>the</strong>lial nests representing remnants <strong>of</strong> <strong>the</strong> dentallamina.Radiologically, a lot <strong>of</strong> jaw diseases associated withunerupted teeth may have an appearance similar to that<strong>of</strong> a dentigerous cyst. Histologic examination, however,will be decisive in ruling out <strong>the</strong>se possibilities amongFig. 4.6. Lining <strong>of</strong> a dentigerous cyst mainly composed <strong>of</strong> mucouscellswhich keratocyst <strong>and</strong> unicystic ameloblastoma (seeSects. 4.3.2.4 <strong>and</strong> 4.4.1.1) are <strong>the</strong> most prevalent. Moreover,<strong>the</strong> radiologic picture <strong>of</strong> <strong>the</strong> dentigerous cyst maybe mimicked by hyperplasia <strong>of</strong> <strong>the</strong> dental follicle, <strong>the</strong>connective tissue capsule that surrounds <strong>the</strong> uneruptedtooth [33].Fibromyxomatous areas in <strong>the</strong> connective tissue wall<strong>of</strong> <strong>the</strong> dentigerous cyst may resemble <strong>the</strong> odontogenicmyxoma (see Sect. 4.4.2.1). The presence <strong>of</strong> odontogenicepi<strong>the</strong>lial rests may lead to <strong>the</strong> erroneous diagnosis<strong>of</strong> one or ano<strong>the</strong>r type <strong>of</strong> epi<strong>the</strong>lial odontogenic tumour[71]. However, identification <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lial cyst liningwill rule out <strong>the</strong>se alternatives.In most instances, dentigerous cysts are a fortuitousfinding on oral radiographs. Only when excessively largemay <strong>the</strong>y cause swelling <strong>of</strong> <strong>the</strong> involved part <strong>of</strong> <strong>the</strong> jaw.If <strong>the</strong>re is inflammation, <strong>the</strong>y will cause pain <strong>and</strong> swelling.Removal <strong>of</strong> <strong>the</strong> cyst wall <strong>and</strong> <strong>the</strong> tooth involved willyield a permanent cure.The eruption cyst is a specific type <strong>of</strong> dentigerous cystlocated in <strong>the</strong> gingival s<strong>of</strong>t tissues overlying <strong>the</strong> crown


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 107<strong>of</strong> an erupting tooth. Mostly, <strong>the</strong>se cysts are short-lived,rupturing with <strong>the</strong> progressive eruption <strong>of</strong> <strong>the</strong> associatedtooth. They are lined by squamous epi<strong>the</strong>lium that isthickened due to inflammatory changes in <strong>the</strong> underlyingconnective tissue <strong>and</strong> thus similar to <strong>the</strong> lining <strong>of</strong> aradicular cyst.4.3.2.2 Lateral Periodontal CystLateral periodontal cysts are rare lesions, derived fromodontogenic epi<strong>the</strong>lial remnants, <strong>and</strong> occurring on <strong>the</strong>lateral aspect or between <strong>the</strong> roots <strong>of</strong> vital teeth (Fig. 4.3)[166]. They are lined by a thin, non-keratinising squamousor cuboidal epi<strong>the</strong>lium with focal, plaque-likethickenings consisting <strong>of</strong> clear cells that may containglycogen (Fig. 4.7) [150].Lateral periodontal cysts do not cause any symptoms.They are fortuitous findings on radiographs where <strong>the</strong>ypresent as a well-demarcated radiolucency on <strong>the</strong> lateralsurface <strong>of</strong> a tooth root. Simple enucleation is adequatetreatment.The botryoid odontogenic cyst represents a multilocularform <strong>of</strong> <strong>the</strong> lateral periodontal cyst [54]. Treatmentby curettage is <strong>the</strong> most appropriate treatment, but recurrencesmay occur [52].Fig. 4.7. Lateral periodontal cyst. The epi<strong>the</strong>lial lining formsplaques consisting <strong>of</strong> clear cells4.3.2.3 Gl<strong>and</strong>ular Odontogenic CystThe gl<strong>and</strong>ular odontogenic cyst , also called sialo-odontogeniccyst is a rare cystic lesion characterised by an epi<strong>the</strong>liallining with cuboidal or columnar cells both at <strong>the</strong>surface <strong>and</strong> lining crypts or cyst-like spaces within <strong>the</strong>thickness <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium [34, 73].The lining epi<strong>the</strong>lium is partly non-keratinising,squamous <strong>and</strong> with focal thickenings similar to <strong>the</strong>plaques in <strong>the</strong> lateral periodontal cyst <strong>and</strong> <strong>the</strong> botryoidodontogenic cyst. There may be a surface layer <strong>of</strong>eosinophilic cuboidal or columnar cells that can havecilia <strong>and</strong> may form papillary projections. Some superficialcells assume an apocrine appearance. Also, mucus-producingcells may be present. Focally, <strong>the</strong> epi<strong>the</strong>liumshows areas <strong>of</strong> increased thickness in which gl<strong>and</strong>ularspaces are formed. Moreover, <strong>the</strong> epi<strong>the</strong>lial cellsmay lie in spherical structures with a whorled appearance(Fig. 4.8).Mucous cells <strong>and</strong> cuboidal cells with cilia may alsooccur in o<strong>the</strong>r jaw cysts, but <strong>the</strong> latter lack <strong>the</strong> o<strong>the</strong>r epi<strong>the</strong>lialfeatures described above. Mucous cells <strong>and</strong> nonkeratinisingsquamous epi<strong>the</strong>lium also occur in mucoepidermoidcarcinoma [91, 177]. However, epi<strong>the</strong>lialplaques consisting <strong>of</strong> clear cells are not a feature <strong>of</strong> thislatter lesion.The gl<strong>and</strong>ular odontogenic cyst most commonly affects<strong>the</strong> body <strong>of</strong> <strong>the</strong> m<strong>and</strong>ible <strong>and</strong> <strong>the</strong> most prominentFig. 4.8. Epi<strong>the</strong>lial lining <strong>of</strong> gl<strong>and</strong>ular odontogenic cyst showingintraepi<strong>the</strong>lial duct formation <strong>and</strong> apocrine differentiation at <strong>the</strong>surfacesymptom is painless swelling [126]. Treatment may beconservative, but recurrence may occur [48].4.3.2.4 Odontogenic KeratocystOdontogenic keratocyst , formerly also called primordialcyst , is defined by <strong>the</strong> presence <strong>of</strong> an epi<strong>the</strong>lial lining notexceeding 10 cell layers in thickness, palisading <strong>of</strong> <strong>the</strong>basal cells, <strong>and</strong> a parakeratinised, corrugated surface[114].Odontogenic keratocysts are common lesions [147,148, 149]. They show a wide age range with a peak frequencyin <strong>the</strong> 2nd <strong>and</strong> 3rd decades, are more commonin males <strong>the</strong>n in females <strong>and</strong> occur twice as frequently in<strong>the</strong> m<strong>and</strong>ible as in <strong>the</strong> maxilla. Involvement <strong>of</strong> <strong>the</strong> gingivals<strong>of</strong>t tissues has also been reported [26]. They alsooccur in cases <strong>of</strong> nevoid basal cell carcinoma syndrome<strong>and</strong> in patients with Marfan’s syndrome [11, 50, 186].


108 P.J. Slootweg4Fig. 4.9. Epi<strong>the</strong>lial lining <strong>of</strong> a keratocyst. The basal palisading<strong>and</strong> <strong>the</strong> corrugated parakeratinised surface are unique to this lesion.Moreover, <strong>the</strong>re is a striking parallel between <strong>the</strong> basal layer<strong>and</strong> <strong>the</strong> surfaceFig. 4.10. In <strong>the</strong> event <strong>of</strong> inflammation, <strong>the</strong> epi<strong>the</strong>lial lining <strong>of</strong> akeratocyst loses its typical pattern to transform into a lattice <strong>of</strong>spongiotic squamous epi<strong>the</strong>liumThe odontogenic keratocyst shows a thin connectivetissue wall lined by stratified squamous epi<strong>the</strong>liumwith a well-defined basal layer <strong>of</strong> palisading columnaror cuboidal cells <strong>and</strong> with a surface <strong>of</strong> a corrugatedlayer <strong>of</strong> parakeratin (Fig. 4.9). Mitotic figurescan be identified in parabasilar <strong>and</strong> midspinous areas[12]. Rushton bodies similar to those seen in radicularcysts may also be present. The underlying cyst wallmay contain tiny daughter cysts <strong>and</strong> solid epi<strong>the</strong>lialnests. Also, epi<strong>the</strong>lial proliferations similar to ameloblastomahave been reported. Daughter cysts <strong>and</strong> intramuralepi<strong>the</strong>lial nests are more common in cystsassociated with <strong>the</strong> nevoid basal cell carcinoma syndrome[185]. When inflamed, <strong>the</strong> odontogenic keratocystloses its typical histologic features, but showsa non-keratinising stratified epi<strong>the</strong>lium exhibitingspongiosis <strong>and</strong> elongated rete pegs supported by a connectivetissue containing a mixed inflammatory infiltrate(Fig. 4.10). Rarely, odontogenic keratocysts showdevelopment <strong>of</strong> epi<strong>the</strong>lial dysplasia <strong>and</strong> squamous cellcarcinoma [87].Immunohistochemical studies have not yielded data<strong>of</strong> diagnostic or prognostic significance [148, 149].Odontogenic keratocysts may also contain mucouscells, melanin-producing cells, dentinoid <strong>and</strong> intramuralcartilage [12, 74, 86, 105]. Ciliated cells may be seen,but in maxillary cases, <strong>the</strong>y could also be <strong>the</strong> result <strong>of</strong> acommunication with <strong>the</strong> maxillary sinus [122]. In addition,<strong>the</strong> cyst wall may contain intramural odontogenicepi<strong>the</strong>lial remnants. Occasionally, intraosseous cysts arelined by orthokeratinised epi<strong>the</strong>lium, thus having <strong>the</strong>appearance <strong>of</strong> an epidermoid cyst. Such cysts are knownas orthokeratinised odontogenic cysts. Their differentiationfrom <strong>the</strong> odontogenic keratocyst with parakeratinisationis clinically important as recurrence <strong>of</strong> orthokeratinisedcysts is rare [188]. Differential diagnosiswith unicystic ameloblastoma (see Sect. 4.4.1.1) maybe difficult. Odontogenic keratocyst exhibits a compactspinous layer <strong>and</strong> a corrugated superficial parakeratinlayer, <strong>and</strong> ameloblastoma a spinous layer with intercellularoedema.Keratocysts do not cause symptoms unless concomitantinflammation causes pain <strong>and</strong> swelling. Radiographsmay reveal extensive uni- or multilocular radiolucentlesions that occupy <strong>the</strong> major part <strong>of</strong> <strong>the</strong> jawwithout having caused any appreciable cortical expansion.When <strong>the</strong> odontogenic keratocyst forms part <strong>of</strong><strong>the</strong> nevoid basal cell carcinoma syndrome, patientsmay show any <strong>of</strong> <strong>the</strong> o<strong>the</strong>r features <strong>of</strong> this syndrome[72].Odontogenic keratocysts tend to recur after enucleation[147, 148, 149]. Sometimes, a partial jaw resectionis needed to a provide permanent cure [182]. If associatedwith <strong>the</strong> nevoid basal cell carcinoma syndrome, <strong>the</strong>chance <strong>of</strong> recurrence is even higher [18]. If located in <strong>the</strong>gingiva, <strong>the</strong> behaviour <strong>of</strong> <strong>the</strong> odontogenic keratocyst isless aggressive [65].As <strong>the</strong>re is sufficient evidence that this lesion actuallyrepresents a cystic neoplasm, <strong>the</strong> most recent WHOclassification proposes <strong>the</strong> diagnostic designation keratocysticodontogenic tumour [181].4.3.2.5 Gingival CystGingival cysts are divided in those occurring in adults<strong>and</strong> those in infants. They are located in <strong>the</strong> gingival tissues.Gingival cysts <strong>of</strong> adults are rarely larger than 1 cm<strong>and</strong> may be multiple. They are lined by ei<strong>the</strong>r thin epi<strong>the</strong>lium<strong>of</strong> one to three cell layers or thicker <strong>and</strong> exhibitkeratinisation. Plaques similar to those occurring in <strong>the</strong>lateral periodontal cyst (see Sect. 4.3.2.2) may be seen[107].


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 109Gingival cysts <strong>of</strong> infants occur ei<strong>the</strong>r singularly ormultiply on <strong>the</strong> edentulous alveolar ridge <strong>of</strong> <strong>the</strong> newborninfant. When occurring at <strong>the</strong> midline <strong>of</strong> <strong>the</strong> palate,<strong>the</strong>y are known as palatal cysts <strong>of</strong> infants. These tinylesions, usually not larger than 3 mm, disappear spontaneouslywithin a short time. Histologically, <strong>the</strong>y resembleepidermoid cysts [24, 94]. Historically, Epstein’spearls <strong>and</strong> Bohn’s nodules are terms that have been usedfor <strong>the</strong>se lesions.4.3.3 Non-Odontogenic Cysts4.3.3.1 Nasopalatine Duct CystNasopalatine duct cysts arise within <strong>the</strong> nasopalatinecanal from epi<strong>the</strong>lial remnants <strong>of</strong> <strong>the</strong> nasopalatine duct.Radiologically, <strong>the</strong>y present as radiolucent lesions situatedbetween <strong>the</strong> roots <strong>of</strong> both maxillary central incisorteeth. The cyst lining may be pseudostratified columnarciliated epi<strong>the</strong>lium, stratified squamous epi<strong>the</strong>lium,columnar or cuboidal epi<strong>the</strong>lium <strong>and</strong> combinations <strong>of</strong><strong>the</strong>se. As surgical treatment comprises emptying <strong>the</strong>nasopalatine canal, <strong>the</strong> specimen always includes <strong>the</strong>artery <strong>and</strong> nerve that run in this anatomic structure.These are seen within <strong>the</strong> fibrous cyst wall <strong>and</strong> form<strong>the</strong> most convincing diagnostic feature, as <strong>the</strong> specificepi<strong>the</strong>lial structures may be obscured by inflammatorychanges. Recurrences are rarely seen, <strong>and</strong> are probablydue to incomplete removal [168].4.3.3.2 Nasolabial CystNasolabial cysts are located in <strong>the</strong> s<strong>of</strong>t tissue just lateralto <strong>the</strong> nose at <strong>the</strong> buccal aspect <strong>of</strong> <strong>the</strong> maxillary alveolarprocess <strong>and</strong> are thought to arise from <strong>the</strong> nasolacrimalduct. Non-ciliated pseudostratified columnar epi<strong>the</strong>liuminterspersed with mucous cells form <strong>the</strong> epi<strong>the</strong>liallining. These features may be lost through squamousmetaplasia [180]. Apocrine metaplasia <strong>of</strong> <strong>the</strong> cyst lininghas also been reported [83]. Treatment consists <strong>of</strong>enucleation.4.3.3.3 Surgical Ciliated CystSurgical ciliated cysts arise from detached portions<strong>of</strong> <strong>the</strong> mucosa that line <strong>the</strong> maxillary antrum <strong>and</strong> areburied within <strong>the</strong> maxillary bone. This may occur aftertrauma or surgical intervention in this area [93]. Mostly,<strong>the</strong> cyst is an incidental radiographic finding, observedas a well-defined unilocular radiolucency adjacent to <strong>the</strong>maxillary antrum.The cyst lining is similar to <strong>the</strong> normal mucosal surface<strong>of</strong> <strong>the</strong> paranasal cavities: pseudostratified ciliatedcolumnar epi<strong>the</strong>lium with interspersed mucous cells.Treatment consists <strong>of</strong> simple enucleation.4.3.4 Pseudocysts4.3.4.1 Solitary Bone CystThe solitary bone cyst , also known as traumatic bone cystor simple bone cyst is confined to <strong>the</strong> m<strong>and</strong>ibular body.Its pathogenesis is ill-understood; a remnant <strong>of</strong> intraosseoushaemorrhage is <strong>the</strong> most favoured hypo<strong>the</strong>sis. Radiographsshow a cavity that varies from less than 1 cmin diameter to one that occupies <strong>the</strong> entire m<strong>and</strong>ibularbody <strong>and</strong> ramus. At surgical exploration, one encountersa fluid-filled cavity. Material for histologic examinationmay be difficult to obtain as a s<strong>of</strong>t tissue lining<strong>of</strong> <strong>the</strong> bony cavity may be entirely absent or very thin.If present, it usually consists only <strong>of</strong> loose fibrovasculartissue, although it may also contain granulation tissuewith signs <strong>of</strong> previous haemorrhage such as cholesterolclefts <strong>and</strong> macrophages loaded with iron pigment [136].Sometimes, this cyst develops simultaneously with a variety<strong>of</strong> fibro-osseous cemental lesions [62].4.3.4.2 Focal Bone Marrow DefectThe focal bone marrow defect represents an asymptomaticradiolucent lesion <strong>of</strong> <strong>the</strong> jaws that containsnormal hematopoietic <strong>and</strong> fatty bone marrow. It is alsocalled osteoporotic bone marrow defect . This conditionis mostly seen at <strong>the</strong> angle <strong>of</strong> <strong>the</strong> m<strong>and</strong>ible where it revealsits presence as a radiolucency with more or lesswell-defined borders. Due to <strong>the</strong> lack <strong>of</strong> radiographicspecificity, <strong>the</strong> lesion is usually biopsied. Then, histologicexamination will reveal <strong>the</strong> presence <strong>of</strong> normalhematopoietic marrow [141]. Of course, fur<strong>the</strong>r treatmentis superfluous.4.4 Odontogenic TumoursOdontogenic tumours comprise a group <strong>of</strong> lesions thathave in common that <strong>the</strong>y arise from <strong>the</strong> odontogenictissue. They develop from <strong>the</strong> epi<strong>the</strong>lial part <strong>of</strong> <strong>the</strong> toothgerm, <strong>the</strong> ectomesenchymal part or from both. Theirbehaviour varies from frankly neoplastic, includingmetastatic potential, to non-neoplastic hamartomatous.Some <strong>of</strong> <strong>the</strong>m may recapitulate normal tooth developmentincluding <strong>the</strong> formation <strong>of</strong> dental hard tissues suchas enamel, dentin <strong>and</strong> cementum [129]. Table 4.2 gives


110 P.J. SlootwegTable 4.2. Odontogenic tumours [73, 181]4Epi<strong>the</strong>lialMesenchymalMixed epi<strong>the</strong>lial <strong>and</strong> mesenchymalMalignantAmeloblastomaCalcifying epi<strong>the</strong>lial odontogenic tumourAdenomatoid odontogenic tumourSquamous odontogenic tumourOdontogenic myxomaOdontogenic fibromaCementoblastomaAmeloblastic fibromaAmeloblastic fibro-odontomaOdontoma – complex typeOdontoma – compound typeCalcifying odontogenic cyst (calcifying cystic odontogenic tumour/dentinogenic ghost cell tumour [181])Odonto-ameloblastomaMalignant ameloblastomaAmeloblastic carcinomaPrimary intraosseous carcinomaClear cell odontogenic carcinomaMalignant epi<strong>the</strong>lial odontogenic ghost cell tumourOdontogenic sarcomaan overview <strong>of</strong> <strong>the</strong> various entities encompassed underthis heading.4.4.1 Odontogenic Tumours –Epi<strong>the</strong>lialEpi<strong>the</strong>lial odontogenic tumours are supposed to bederived from odontogenic epi<strong>the</strong>lium: dental lamina,enamel organ <strong>and</strong> Hertwig’s root sheath. As <strong>the</strong>re is nocontribution, ei<strong>the</strong>r proliferative or inductive, from <strong>the</strong>odontogenic mesenchyme, <strong>the</strong>se lesions do not containdental hard tissues or myxoid tissue resembling <strong>the</strong> dentalpapilla.4.4.1.1 AmeloblastomaICD-O:9310/0Ameloblastomas closely resemble <strong>the</strong> epi<strong>the</strong>lial part <strong>of</strong><strong>the</strong> tooth germ. They behave aggressively locally, but donot metastasise.It is <strong>the</strong> most common odontogenic tumour [34] <strong>and</strong>may occur at any age, although cases in <strong>the</strong> first decadeare rare. Maxillary cases are far outnumbered by m<strong>and</strong>ibularones. Rarely, <strong>the</strong> sinonasal cavities are involved[130, 139].The intraosseous lesions are solid, solid with cysticparts, multicystic or unicystic. In <strong>the</strong> gingiva, <strong>the</strong> tumourshave a white fibrous appearance on <strong>the</strong> cut surface,due to <strong>the</strong> preponderance <strong>of</strong> fibrous stroma <strong>of</strong> lesionsat this site.Ameloblastomas consist <strong>of</strong> ei<strong>the</strong>r anastomosing epi<strong>the</strong>lialstr<strong>and</strong>s <strong>and</strong> fields or discrete epi<strong>the</strong>lial isl<strong>and</strong>s.The former pattern is called <strong>the</strong> plexiform type, <strong>the</strong> o<strong>the</strong>r<strong>the</strong> follicular (Figs 4.11, 4.12). Both may occur withinone <strong>and</strong> <strong>the</strong> same lesion [73, 181]. The peripheral cells at<strong>the</strong> border with <strong>the</strong> adjacent fibrous stroma are columnarwith nuclei usually in <strong>the</strong> apical half <strong>of</strong> <strong>the</strong> cell bodyaway from <strong>the</strong> basement membrane. The cells lying morecentrally are fusiform to polyhedral <strong>and</strong> loosely connectedto each o<strong>the</strong>r through cytoplasmic extensions. Especiallyin <strong>the</strong> follicular type, an increase in intercellularoedema may cause cysts that coalesce to form <strong>the</strong> largecavities responsible for <strong>the</strong> multicystic gross appearanceameloblastomas may show. In <strong>the</strong> plexiform type, cystformation is usually <strong>the</strong> result <strong>of</strong> stromal degeneration.Condensation <strong>of</strong> collagenous fibres may cause a juxtaepi<strong>the</strong>lialeosinophilic hyaline b<strong>and</strong>. At <strong>the</strong> periphery<strong>of</strong> <strong>the</strong> lesion, <strong>the</strong> tumour infiltrates <strong>the</strong> adjacent cancellousbone. The lower cortical border <strong>of</strong> <strong>the</strong> m<strong>and</strong>ible <strong>and</strong><strong>the</strong> periosteal layer usually exp<strong>and</strong>, but will not be perforated,<strong>the</strong> periosteum in particular forming a barrier[99]. Spread into s<strong>of</strong>t tissues is highly unusual; whenobserved, it is probably an ameloblastic carcinoma, a lesionto be discussed later on (see Sect. 4.4.4.2). Mitoticfigures may occur within <strong>the</strong> peripheral columnar aswell as in <strong>the</strong> stellate reticulum-like cells. In <strong>the</strong> absence<strong>of</strong> cytonuclear atypia <strong>and</strong> with a normal configuration,<strong>the</strong>y are without prognostic significance.


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 111Fig. 4.11. Large epi<strong>the</strong>lial areas <strong>of</strong> loosely structured spindle epi<strong>the</strong>liumenclosing liquefacting stromal areas are typical <strong>of</strong> a plexiformameloblastoma. The epi<strong>the</strong>lial cells facing <strong>the</strong> stroma showpalisadingFig. 4.13. Desmoplastic ameloblastoma consists <strong>of</strong> denselypacked spindle cells lying in a fibrous stroma. Palisading <strong>of</strong> peripheralcells is not a conspicuous feature in this type <strong>of</strong> ameloblastomaFig. 4.12. In cases <strong>of</strong> follicular ameloblastoma, <strong>the</strong> tumour consists<strong>of</strong> epi<strong>the</strong>lial isl<strong>and</strong>s with a loose oedematous centre <strong>and</strong> a peripheralrim <strong>of</strong> palisading cells. Liquefaction <strong>of</strong> <strong>the</strong>ir centre resultsin cyst formationFig. 4.14. In unicystic ameloblastoma, <strong>the</strong> tumour consists <strong>of</strong>cyst-lining epi<strong>the</strong>lium that still shows <strong>the</strong> typical features <strong>of</strong> ameloblastoma:loose epi<strong>the</strong>lium <strong>and</strong> a rim <strong>of</strong> palisading cells facing<strong>the</strong> stromaAcanthomatous <strong>and</strong> granular cell type ameloblastomaare variants <strong>of</strong> follicular ameloblastoma with squamousmetaplasia <strong>and</strong> granular cells respectively. If keratinisationis abundant, leading to large cavities filledwith keratin, lesions are called keratoameloblastoma[135]. In <strong>the</strong>se tumours acantholysis may lead to a pseudopapillarylining that characterises <strong>the</strong> variant calledpapilliferous keratoameloblastoma.The basal cell ( basaloid) ameloblastoma is composed<strong>of</strong> nests <strong>of</strong> basaloid cells with a peripheral rim <strong>of</strong> cuboidalcells <strong>and</strong> does not display a well-developed, loose oedematouscentre.Desmoplastic ameloblastoma shows a dense collagenousstroma, <strong>the</strong> epi<strong>the</strong>lial component being reduced tonarrow, compressed str<strong>and</strong>s <strong>of</strong> epi<strong>the</strong>lium. When <strong>the</strong>sestr<strong>and</strong>s broaden to form larger isl<strong>and</strong>s, a peripheral rim<strong>of</strong> dark staining cuboidal cells <strong>and</strong> a compact centre inwhich spindle-shaped epi<strong>the</strong>lial cells assume a whorlingpattern may be discerned (Fig. 4.13). Within <strong>the</strong> stromalcomponent, active bone formation can be observed[119].Unicystic ameloblastoma represents a cyst that islined by ameloblastomatous epi<strong>the</strong>lium (Fig. 4.14) [115].This epi<strong>the</strong>lium may proliferate to form intraluminalnodules with <strong>the</strong> architecture <strong>of</strong> plexiform ameloblastoma.Downward proliferation <strong>of</strong> this epi<strong>the</strong>lium may leadto infiltration <strong>of</strong> <strong>the</strong> fibrous cyst wall by ameloblastomanests. Sometimes, <strong>the</strong> cyst lining itself lacks any featuresindicative <strong>of</strong> ameloblastoma, <strong>the</strong>se being confined to intramuralepi<strong>the</strong>lial nests [47]. Inflammatory alterationsmay obscure <strong>the</strong> specific histologic details to such an extentthat none are left.Ameloblastomas may also contain clear cells as wellas mucous cells [100, 184].Epi<strong>the</strong>lial nests resembling ameloblastoma may befound in calcifying odontogenic cysts <strong>and</strong> ameloblas-


112 P.J. Slootweg4tic fibromas, lesions to be discussed under <strong>the</strong> appropriateheadings (Sects. 4.4.3.1 <strong>and</strong> 4.4.3.6). Also, epi<strong>the</strong>lialnests in <strong>the</strong> dental follicle that surrounds an impactedtooth <strong>and</strong> in <strong>the</strong> wall <strong>of</strong> odontogenic cysts may mimicameloblastoma. Maxillary ameloblastomas may bemistaken for solid-type adenoid cystic carcinomas (seeChap. 5).Ameloblastomas usually have swelling as <strong>the</strong> mostprominent symptom. In <strong>the</strong> maxilla, growth into <strong>the</strong>paranasal sinuses allows tumours to attain a considerablesize without causing any external deformity. Radiographically,ameloblastoma is a radiolucent lesion that isusually multilocular, <strong>the</strong> so-called soap bubble appearance,or unilocular with scalloped outlines [130].Sometimes, ameloblastomas present as s<strong>of</strong>t tissueswellings occurring in <strong>the</strong> tooth-bearing areas <strong>of</strong> <strong>the</strong>maxilla or m<strong>and</strong>ible without involvement <strong>of</strong> <strong>the</strong> underlyingbone. This peripheral ameloblastoma should not beconfused with intraosseous ameloblastomas that spreadfrom within <strong>the</strong> jaw into <strong>the</strong> overlying gingiva [117]. In<strong>the</strong> past, <strong>the</strong>se lesions have also been described as odontogenicgingival epi<strong>the</strong>lial hamartoma [6].Clinically, some variants differ slightly from <strong>the</strong> prototypicameloblastoma. Desmoplastic ameloblastomaoccurs more <strong>of</strong>ten in <strong>the</strong> anterior parts <strong>of</strong> both maxilla<strong>and</strong> m<strong>and</strong>ible than <strong>the</strong> o<strong>the</strong>r types, which favour <strong>the</strong>posterior m<strong>and</strong>ible [119]. Unicystic ameloblastoma occursat a lower mean age than <strong>the</strong> o<strong>the</strong>r types <strong>and</strong> <strong>of</strong>tenhas a radiographic appearance similar to a dentigerouscyst because <strong>of</strong> its association with an impactedtooth [115].Treatment <strong>of</strong> ameloblastoma consists <strong>of</strong> adequate tumourremoval including a margin <strong>of</strong> uninvolved tissue.For peripheral ameloblastoma simple excision willbe sufficient treatment [47, 115]. For unicystic ameloblastomawith <strong>the</strong> ameloblastomatous epi<strong>the</strong>lium confinedto <strong>the</strong> cyst lining, enucleation is adequate <strong>the</strong>rapy,but in cases <strong>of</strong> intramural proliferation, treatmentshould be <strong>the</strong> same as for <strong>the</strong> o<strong>the</strong>r ameloblastoma types[133]. When adequately treated, ameloblastomas are notexpected to recur. Adequate removal, however, may bedifficult to realise in maxillary cases that grow posterocranially.In that case, extension into <strong>the</strong> cranial cavitymay be fatal [103]. In rare instances, metastatic deposits,mainly to <strong>the</strong> lung, have been observed. Lesions showingthis behaviour are called malignant ameloblastoma(see Sect. 4.4.4.1).4.4.1.2 Calcifying Epi<strong>the</strong>lialOdontogenic TumourICD-O:9340/0The calcifying epi<strong>the</strong>lial odontogenic tumour , alsonamed Pindborg tumour occurs between <strong>the</strong> 2nd <strong>and</strong>6th decade <strong>and</strong> mainly involves <strong>the</strong> posterior jaw area.Also, cases located at <strong>the</strong> gingiva may be seen [63,116].The tumour consists <strong>of</strong> sheets <strong>of</strong> polygonal cells withample eosinophilic cytoplasm, distinct cell borders <strong>and</strong>very conspicuous intercellular bridges. Nuclei are pleomorphicwith prominent nucleoli; cells with giant nuclei<strong>and</strong> multiple nuclei are also present (Fig. 4.15). Mitoticfigures, however, are absent. Clear cell differentiationmay occur [58]. The epi<strong>the</strong>lial tumour isl<strong>and</strong>s as well as<strong>the</strong> surrounding stroma frequently contain concentricallylamellated calcifications. The stroma contains eosinophilicmaterial that stains like amyloid (Fig. 4.16)[73, 181]. The presence <strong>of</strong> bone <strong>and</strong> cementum in <strong>the</strong> tumourhas also been reported [155]. There is no encapsulation.The tumour grows into <strong>the</strong> cancellous spaces <strong>of</strong><strong>the</strong> adjacent jaw bone while causing expansion <strong>and</strong> thinning<strong>of</strong> <strong>the</strong> cortical bone.Due to its pronounced nuclear pleomorphism, <strong>the</strong> tumourmay be mistaken for a high-grade malignant carcinoma;<strong>the</strong> absence <strong>of</strong> mitotic figures should preventthis diagnostic error.Swelling is <strong>the</strong> most common clinical symptom <strong>of</strong>this tumour. Radiographically, <strong>the</strong> tumour is characterisedby a diffuse mixed radiodense <strong>and</strong> radiolucent appearance.Quite <strong>of</strong>ten, an unerupted tooth lies buriedin <strong>the</strong> tumour mass. Surgery consists <strong>of</strong> removal with amargin <strong>of</strong> uninvolved tissue. Recurrences are occasionallyseen, in particular with <strong>the</strong> clear cell variant [58].Cases occurring in <strong>the</strong> extragnathic gingival tissue canbe treated by simple excision as <strong>the</strong>y are less aggressivethan <strong>the</strong> intraosseous ones [63].Metastatic disease is only seen in cases that combine<strong>the</strong> appearance <strong>of</strong> a calcifying epi<strong>the</strong>lial odontogenic tumourwith <strong>the</strong> presence <strong>of</strong> mitotic activity, suggestingmalignant transformation [175]. Mitotic activity has alsobeen seen in combination with perforation <strong>of</strong> corticalplates <strong>and</strong> invasion <strong>of</strong> blood vessels, both also highly unusualfor calcifying epi<strong>the</strong>lial odontogenic tumours [27].Apparently, mitoses in this tumour indicate malignancy.4.4.1.3 AdenomatoidOdontogenic TumourICD-O:9300/0Adenomatoid odontogenic tumour probably representsan odontogenic hamartoma ra<strong>the</strong>r than a neoplasm[73, 181]. The lesion is mostly is seen in peoplein <strong>the</strong>ir 2nd decade. The anterior maxilla is <strong>the</strong> favouredsite <strong>and</strong> <strong>the</strong> lesion is <strong>of</strong>ten associated with animpacted tooth [120]. Grossly, <strong>the</strong> adenomatoid odontogenictumour is a cyst that embraces <strong>the</strong> crown <strong>of</strong><strong>the</strong> involved tooth.The lesion consists <strong>of</strong> two different cell populations:spindle-shaped <strong>and</strong> columnar. The spindle-shapedcells form whorled nodules that may contain drop-


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 113Fig. 4.15. Nuclear atypia, ample cytoplasm <strong>and</strong> pronounced intercellularbridging are typical <strong>of</strong> a calcifying epi<strong>the</strong>lial odontogenictumourFig. 4.17. Areas <strong>of</strong> small spindle cells <strong>and</strong> cylindrical cells facingextracellular material characterise <strong>the</strong> adenomatoid odontogenictumourOdontogenic adenomatoid tumour usually has swellingat <strong>the</strong> site <strong>of</strong> a missing tooth as a presenting symptom.Radiographically, this missing tooth is seen surroundedby a radiolucency that may contain multipletiny opaque foci.Treatment consists <strong>of</strong> simple enucleation.4.4.1.4 SquamousOdontogenic TumourFig. 4.16. Extracellular material staining for amyloid with congored is ano<strong>the</strong>r characteristic feature <strong>of</strong> a calcifying epi<strong>the</strong>lialodontogenic tumourlets <strong>of</strong> eosinophilic material. A lattice <strong>of</strong> thin epi<strong>the</strong>lialstr<strong>and</strong>s may connect <strong>the</strong>se nodules to each o<strong>the</strong>r.The columnar cells line duct-like spaces with a lumenei<strong>the</strong>r empty or containing eosinophilic material <strong>and</strong>may form curvilinear opposing rows with interposedeosinophilic material (Fig. 4.17). In <strong>the</strong> stroma, <strong>the</strong>reare large aggregates <strong>of</strong> eosinophilic hyaline material,which is judged to be a dysplastic form <strong>of</strong> dentin, orcementum, or to be a metaplastic reaction <strong>of</strong> <strong>the</strong> stromaltissue [73, 120, 181]. Also, concentrically laminatedcalcified bodies similar to those seen in calcifyingepi<strong>the</strong>lial odontogenic tumours may occur.In some adenomatoid odontogenic tumours, areas<strong>of</strong> eosinophilic cells with well-defined cell boundaries<strong>and</strong> prominent intercellular bridges similar to thoseobserved in <strong>the</strong> calcifying epi<strong>the</strong>lial odontogenic tumourare seen [110]. They do not influence <strong>the</strong> biologicbehaviour <strong>of</strong> this tumour <strong>and</strong> are considered to be part<strong>of</strong> its histologic spectrum as is <strong>the</strong> presence <strong>of</strong> melaninpigment [78, 178].ICD-O:9312/0Squamous odontogenic tumour is a rare lesion thatmainly involves <strong>the</strong> periodontal tissues. There is nopreference for ei<strong>the</strong>r sex or jaw area [73, 181].The lesion is composed <strong>of</strong> isl<strong>and</strong>s <strong>of</strong> well-differentiatedsquamous epi<strong>the</strong>lium surrounded by mature fibrousconnective tissue. There is no cellular atypia.There is spinous differentiation with well-defined intercellularbridges, but keratinisation is unusual. In <strong>the</strong>epi<strong>the</strong>lial isl<strong>and</strong>s, cystic degeneration <strong>and</strong> calcificationmay occur. Invasion into cancellous bone may be present.The absence <strong>of</strong> cytonuclear atypia rules out well-differentiatedsquamous cell carcinoma <strong>and</strong> <strong>the</strong> absence <strong>of</strong>peripheral palisading <strong>of</strong> columnar cells excludes ameloblastomaas an alternative diagnosis. Sometimes, intramuralepi<strong>the</strong>lial proliferation in jaw cysts may simulatesquamous odontogenic tumour [187].The lesion may cause loosening <strong>of</strong> <strong>the</strong> teeth involved.Radiographically, lucent areas are seen. Treatmentconsists <strong>of</strong> conservative removal <strong>of</strong> <strong>the</strong> tumourtissue. Occasionally, more extensive local spread maynecessitate surgical excision with wider margins[142]. Sometimes, multicentric presentation may occur[79].


114 P.J. Slootweg4Fig. 4.18. Odontogenic myxoma is composed <strong>of</strong> poorly cellularmyxoid material that surrounds pre-existing jaw boneFig. 4.19. Odontogenic fibroma consists <strong>of</strong> fibrous areas containingepi<strong>the</strong>lial odontogenic nests4.4.2 Odontogenic Tumours –MesenchymalMesenchymal odontogenic tumours are derived from<strong>the</strong> ectomesenchymal part <strong>of</strong> <strong>the</strong> tissues that participatein <strong>the</strong> development <strong>of</strong> teeth <strong>and</strong> periodontal tissues.Odontogenic epi<strong>the</strong>lial rests may be part <strong>of</strong> <strong>the</strong> histologicpicture <strong>the</strong>y show, but only fortuitously by tumourtissue-engulfed structures. They have no neoplastic orinductive potential.4.4.2.1 Odontogenic MyxomaICD-O:9320/0Odontogenic myxomas usually occur in <strong>the</strong> 2nd or 3rddecade <strong>of</strong> life, although cases occurring at a very youngor old age have been reported. It is one <strong>of</strong> <strong>the</strong> more commonodontogenic tumours [8].Myxomas consist <strong>of</strong> ra<strong>the</strong>r monotonous cells withmultipolar or bipolar slender cytoplasmic extensionsthat lie in a myxoid stroma. Nuclei vary from round t<strong>of</strong>usiform in appearance. Binucleated cells <strong>and</strong> mitoticfigures are present, but scarce (Fig. 4.18). Occasionally,<strong>the</strong> lesion contains odontogenic epi<strong>the</strong>lial rests.They are a fortuitous finding without any diagnosticor prognostic significance. Myxoma cells are positivefor vimentin <strong>and</strong> muscle-specific actin, whereas positivityfor S-100 is controversial [82, 102, 169].Myxoma may be mimicked by dental follicle <strong>and</strong>dental papilla. Both contain myxoid areas [71, 101,165]. Dental papilla tissue can be distinguished frommyxoma by <strong>the</strong> presence <strong>of</strong> a peripheral layer <strong>of</strong> columnarodontoblasts. For both dental papilla <strong>and</strong> dentalfollicle clinical <strong>and</strong> radiographic data are decisivein avoiding misinterpretation <strong>of</strong> myxomatous tissue injaw specimens: in <strong>the</strong> first case, a tooth germ lies in <strong>the</strong>jaw area from which <strong>the</strong> submitted tissue has been takenwhereas in <strong>the</strong> second case, <strong>the</strong> tissue sample covered<strong>the</strong> crown area <strong>of</strong> an impacted tooth.Odontogenic myxomas occur in <strong>the</strong> maxilla as well as<strong>the</strong> m<strong>and</strong>ible <strong>and</strong> in both anterior <strong>and</strong> posterior parts.Swelling may be <strong>the</strong> presenting sign as well as disturbancesin tooth eruption or changes in position <strong>of</strong> teethalready erupted. In maxillary cases, nasal stuffiness maybe <strong>the</strong> presenting sign due to tumour growth in nasal<strong>and</strong> paranasal cavities. Radiographically, lesions show aunilocular or soap-bubble appearance.As <strong>the</strong> lesion lacks encapsulation, treatment usuallyconsists <strong>of</strong> excision with a margin <strong>of</strong> uninvolved tissue[161]. Incidentally, cases with extremely aggressive localgrowth have been reported [35].4.4.2.2 Odontogenic FibromaICD-O:9321/0Odontogenic fibroma is a controversial entity. Uncertaintyexists about <strong>the</strong> broadness <strong>of</strong> histologic spectrumthat <strong>the</strong>se lesions may show, <strong>and</strong> about its distinctionfrom o<strong>the</strong>r fibrous jaw lesions [73, 181].Odontogenic fibroma has an age distribution <strong>of</strong> 9–80 years <strong>and</strong> occurs predominantly in females [55]. Thelesion is seen within <strong>the</strong> jaw as well as in <strong>the</strong> gingiva[32].Odontogenic fibroma consists <strong>of</strong> fibroblasts lyingin a background <strong>of</strong> myxoid material intermingled withcollagen fibres that may vary from delicate to coarse.Odontogenic epi<strong>the</strong>lium, ei<strong>the</strong>r scarce or abundant,may occur (Fig. 4.19). Only rarely is <strong>the</strong> epi<strong>the</strong>lialcomponent so conspicuous that differentiation betweenodontogenic fibroma <strong>and</strong> ameloblastoma maybe difficult [66]. This histologic spectrum may exp<strong>and</strong>to include cell-rich myxoid areas, a greater epi<strong>the</strong>lialcomponent <strong>and</strong> varying amounts <strong>of</strong> amorphous calcifiedglobules or mineralised collagenous matrix. Tu-


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 115mours with this more variegated histology have beenreferred to as complex odontogenic fibroma or WHOtypeodontogenic fibroma [73, 181]. Odontogenic fibromamay also contain granular cells. These lesionshave been called granular cell odontogenic fibromasor, alternatively, granular cell ameloblastic fibromas(see Sect. 4.4.3.1). This tumour, however, could alsorepresent a unique entity: central odontogenic granularcell tumour [15, 170]. The granular cells are negativefor epi<strong>the</strong>lial markers <strong>and</strong> S-100 whereas positivityfor CD68 suggests a histiocytic nature [15, 23].Rarely, this tumour may show atypical histologic featuresincluding mitotic activity <strong>and</strong> aggressive behaviour[121].There are also lesions that combine <strong>the</strong> histologic features<strong>of</strong> giant cell granuloma <strong>and</strong> central odontogenic fibroma[108]. Their aggressive nature suggests that <strong>the</strong>giant cell granuloma (see Sect. 4.6.1) component determines<strong>the</strong> clinical behaviour.When odontogenic fibromas show a preponderance<strong>of</strong> myxoid material, distinguishing <strong>the</strong>m from odontogenicmyxomas may become problematic. It is probablybest to consider such cases to be myxomas <strong>and</strong> to treat<strong>the</strong>m accordingly.Ano<strong>the</strong>r significant diagnostic problem is <strong>the</strong> distinctionbetween odontogenic fibroma <strong>and</strong> desmoplasticfibroma, which is clinically very important, as <strong>the</strong>former is benign whereas <strong>the</strong> latter shows aggressive behaviour[160]. Lesions with features <strong>of</strong> both odontogenicfibroma <strong>and</strong> desmoplastic fibroma may occur in patientswith tuberous sclerosis [10].Peripheral odontogenic fibroma should not be confusedwith peripheral ossifying fibroma, a gingival s<strong>of</strong>ttissue lesion characterised by <strong>the</strong> presence <strong>of</strong> mineralisedmaterial <strong>of</strong> various appearances, but lacking odontogenicepi<strong>the</strong>lium (see Chap. 3) [69]. Peripheral odontogenicfibroma has also to be distinguished from peripheralameloblastoma, <strong>the</strong> former lesion lacking ameloblastomatousepi<strong>the</strong>lium. Cases <strong>of</strong> peripheral odontogenic fibromawith an extensive epi<strong>the</strong>lial component have in<strong>the</strong> past been reported with <strong>the</strong> designation odontogenicepi<strong>the</strong>lial hamartoma [176].All histologic features shown by odontogenic fibromamay also be displayed by <strong>the</strong> dental follicle [37, 71, 85,165]. In <strong>the</strong>se cases, <strong>the</strong> radiographic appearance <strong>of</strong> <strong>the</strong>lesion, a small radiolucent rim surrounding <strong>the</strong> crown<strong>of</strong> a tooth buried within <strong>the</strong> jaw, will make <strong>the</strong> distinction.Central odontogenic fibromas may present as localbony expansions <strong>of</strong> <strong>the</strong> involved jaw area. Quite <strong>of</strong>ten,<strong>the</strong>y are incidental findings on radiographs performedfor o<strong>the</strong>r diagnostic purposes: demarcated unilocularradiolucencies located adjacent to <strong>the</strong> roots <strong>of</strong> <strong>the</strong> neighbouringteeth or surrounding an impacted tooth. Peripheralodontogenic fibromas are firm-elastic gingivalswellings.Fig. 4.20. A hard tissue mass firmly connected to <strong>the</strong> root surface<strong>of</strong> <strong>the</strong> tooth involved is diagnostic for cementoblastomaTreatment consists <strong>of</strong> enucleation. Peripheral cases,however, may recur after excision [32].4.4.2.3 CementoblastomaICD-O:9273/0Cementoblastomas are heavily mineralised cementummasses connected to <strong>the</strong> apical root part <strong>of</strong> a tooth(Fig. 4.20) [73, 181]. These tumours are most <strong>of</strong>ten seenin young adults <strong>and</strong> show a predilection for males [14].They are composed <strong>of</strong> a vascular, loose-textured fibroustissue that surrounds coarse trabeculae <strong>of</strong> basophilicmineralised material bordered by plump cellswith ample cytoplasm <strong>and</strong> large but not atypical nuclei.Mitotic figures are rare. At <strong>the</strong> periphery, <strong>the</strong> mineralisedmaterial may form radiating spikes. Also, osteoclasticgiant cells form part <strong>of</strong> <strong>the</strong> histologic spectrum.The hard tissue component is connected with <strong>the</strong> root<strong>of</strong> <strong>the</strong> involved tooth, which usually shows signs <strong>of</strong> externalresorption. The sharp border between <strong>the</strong> tubulardentin <strong>of</strong> <strong>the</strong> root <strong>and</strong> <strong>the</strong> hard tissue component forms<strong>the</strong> hallmark <strong>of</strong> cementoblastomas (Fig. 4.21).All features <strong>of</strong> <strong>the</strong> cementoblastoma may also beshown by <strong>the</strong> osteoblastoma, except <strong>the</strong> connection with<strong>the</strong> tooth root. Therefore, cases in which this connectioncannot be demonstrated should be diagnosed as osteoblastoma<strong>and</strong> not cementoblastoma [156].


116 P.J. Slootweg4Fig. 4.21. Higher magnification <strong>of</strong> Fig. 4.20 shows <strong>the</strong> continuitybetween <strong>the</strong> root forming tubular dentin (right side) <strong>and</strong> <strong>the</strong> cementalmasses <strong>of</strong> <strong>the</strong> cementoblastomaPain is <strong>the</strong> most common presenting symptom <strong>and</strong><strong>the</strong> posterior jaw areas are <strong>the</strong> predilection site. Sometimes,<strong>the</strong> lesion is attached to multiple neighbouringteeth [156]. Radiographically, <strong>the</strong> lesion is demarcatedwith a mixed lucent <strong>and</strong> dense appearance <strong>and</strong> is continuouswith <strong>the</strong> partially resorbed root <strong>of</strong> a tooth. As recurrence<strong>and</strong> continued growth are possible, treatmentshould consist <strong>of</strong> removal <strong>of</strong> <strong>the</strong> lesion along with <strong>the</strong> affectedtooth or teeth, <strong>and</strong> should also include some adjacentjaw bone [14].4.4.3. Odontogenic Tumours –Mixed Epi<strong>the</strong>lial<strong>and</strong> MesenchymalMixed odontogenic tumours are composed <strong>of</strong> both epi<strong>the</strong>lial-derived<strong>and</strong> mesenchymal-derived tissues. Thesetumours recapitulate proliferation <strong>and</strong> differentiation asseen in <strong>the</strong> developing teeth. Deposition <strong>of</strong> <strong>the</strong> dental hardtissues – enamel <strong>and</strong> dentin – may also occur [73, 174,181]. Lesions with an identical histology can show neoplasticas well as hamartomatous behaviour [118, 153].4.4.3.1 Ameloblastic FibromaICD-O:9330/0Ameloblastic fibroma lacks a hard tissue component,only displaying s<strong>of</strong>t tissues similar to those found in <strong>the</strong>immature tooth germ.It is one <strong>of</strong> <strong>the</strong> less common lesions [34, 109, 127].Mean age <strong>of</strong> occurrence is 14.8 years, but cases maybe seen from as young as only 7 weeks up to as old as62 years. Most cases are seen in <strong>the</strong> posterior m<strong>and</strong>ible[96, 118].The epi<strong>the</strong>lial part <strong>of</strong> ameloblastic fibroma consists<strong>of</strong> branching <strong>and</strong> anastomosing epi<strong>the</strong>lial str<strong>and</strong>sthat form knots <strong>of</strong> varying size. These knots have a peripheralrim <strong>of</strong> columnar cells that embraces a looselyarranged spindle-shaped epi<strong>the</strong>lium. These epi<strong>the</strong>lialstr<strong>and</strong>s lie in a myxoid cell-rich mesenchyme. Theamount <strong>of</strong> epi<strong>the</strong>lium may vary among cases <strong>and</strong> regionallywithin an individual case. There is no formation<strong>of</strong> dental hard tissues. Mitotic figures, ei<strong>the</strong>r in epi<strong>the</strong>liumor mesenchyme, are extremely rare; when easilyfound, <strong>the</strong>y should raise concern about <strong>the</strong> benign nature<strong>of</strong> <strong>the</strong> case.Ameloblastic fibroma may contain granular cells.Whe<strong>the</strong>r <strong>the</strong>se lesions should be called granular cell ameloblasticfibroma or granular cell odontogenic fibroma iscontroversial (see also Sect. 4.4.2.2) [118, 170].The epi<strong>the</strong>lial component <strong>of</strong> ameloblastic fibromaclosely resembles that <strong>of</strong> ameloblastoma. The stromalcomponent, however, is entirely different: in ameloblastomait is mature fibrous connective tissue whereasin <strong>the</strong> ameloblastic fibroma it is immature, embryonic,cell-rich myxoid tissue. Areas similar to ameloblasticfibroma may also be observed in <strong>the</strong> hyperplastic dentalfollicle [71, 165]. The radiographic appearance makes<strong>the</strong> distinction; a radiolucent rim surrounding an uneruptedtooth in <strong>the</strong> case <strong>of</strong> a dental follicle <strong>and</strong> an expansiveradiolucent jaw lesion in <strong>the</strong> case <strong>of</strong> an ameloblasticfibroma.Most cases <strong>of</strong> ameloblastic fibroma present as painlessswellings or are discovered due to disturbances intooth eruption. Radiographically, <strong>the</strong> tumour presentsas a well-demarcated expansive radiolucency, <strong>of</strong>ten inconnection with a malpositioned tooth.Treatment consists <strong>of</strong> enucleation <strong>and</strong> curettage. Insome cases, recurrence may occur [118]. Sometimes,ameloblastic fibroma may progress to malignancy.These lesions are characterised by increased cellularity<strong>and</strong> mitotic activity <strong>of</strong> <strong>the</strong> mesenchymal component<strong>and</strong> <strong>the</strong>refore known as ameloblastic fibrosarcoma (seeSect. 4.4.4.6) [96].4.4.3.2 AmeloblasticFibro-OdontomaICD-O:9290/0Ameloblastic fibro-odontomas are lesions that combinea s<strong>of</strong>t tissue component, similar to ameloblastic fibroma,with <strong>the</strong> presence <strong>of</strong> dentin <strong>and</strong> enamel. In rare cases,only dentin is formed; those tumours are called ameloblasticfibro-dentinoma (ICD-O:9271/0) [73, 181].


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 117as those as already mentioned (see Sect. 4.4.3.1). Ameloblasticfibro-odontomas can be distinguished from ameloblastomasby <strong>the</strong> presence <strong>of</strong> cellular myxoid tissue<strong>and</strong> <strong>of</strong> dentin <strong>and</strong> enamel.Most cases <strong>of</strong> ameloblastic fibro-odontoma presentas painless swelling or are discovered due to disturbancesin tooth eruption. Radiographically, <strong>the</strong> tumourpresents as a well-demarcated expansive radiolucencywith a radiopaque centre. Treatment consists<strong>of</strong> enucleation <strong>and</strong> curettage. Recurrence is rarely seen[46].Fig. 4.22. Ameloblastic fibro-odontoma combines <strong>the</strong> s<strong>of</strong>t tissueelements <strong>of</strong> an ameloblastic fibroma with <strong>the</strong> deposition <strong>of</strong> <strong>the</strong>dental hard tissues enamel <strong>and</strong> dentin. Cavities in <strong>the</strong> homogeneouseosinophilic dentin contain high-columnar ameloblasts lyingdown enamel matrix (deep purple)Fig. 4.23. Panoramic radiograph showing radiodense mass inconnection with a tooth germ, a picture typical <strong>of</strong> complex odontomaAmeloblastic fibro-odontomas are rare [34, 127].They occur primarily within <strong>the</strong> first two decades <strong>and</strong>have <strong>the</strong> posterior jaw areas as sites <strong>of</strong> predilection, <strong>the</strong>m<strong>and</strong>ible more <strong>of</strong>ten involved than <strong>the</strong> maxilla [118,153].The s<strong>of</strong>t tissue component is identical to that <strong>of</strong> ameloblasticfibroma. Dentin may be formed ei<strong>the</strong>r as eosinophilicmineralised material containing tubuli, justas in normal teeth, but it may also form as an homogeneouseosinophilic mass with sparse cells included.It always lies in close association with adjacent epi<strong>the</strong>lium<strong>and</strong> forms <strong>the</strong> scaffold for <strong>the</strong> deposition <strong>of</strong> enamelmatrix that is laid down at <strong>the</strong> epi<strong>the</strong>lial–dentin interfaceby columnar epi<strong>the</strong>lial cells that have reached<strong>the</strong>ir terminal differentiation as ameloblasts. The dentalhard tissues are arranged haphazardly without any reminiscence<strong>of</strong> <strong>the</strong> orderly structure characterising normalteeth (Fig. 4.22).Hyperplastic dental follicles may also show focal areaswith <strong>the</strong> appearance <strong>of</strong> ameloblastic fibro-odontoma.Differential diagnostic considerations are <strong>the</strong> same4.4.3.3 Odontoma –Complex TypeICD-O:9282/0Complex odontoma is a lesion composed <strong>of</strong> a haphazardconglomerate <strong>of</strong> dental hard tissues. This hamartoma isone <strong>of</strong> <strong>the</strong> more common odontogenic lesions [34, 127].The posterior m<strong>and</strong>ible is <strong>the</strong> favoured site (Fig. 4.23).Ages at which this lesion occurs are difficult to determineas lesions may be present unnoticed for a longtime, <strong>the</strong> age distribution mentioned reflecting <strong>the</strong> agewhen <strong>the</strong> lesion was found ra<strong>the</strong>r than <strong>the</strong> age at whichit formed. This explains <strong>the</strong> enormous range in age distributionreported: 2–74 years [118].Complex odontomas consist <strong>of</strong> a usually well-delineatedmass <strong>of</strong> dental hard tissues in a haphazard arrangement.The bulk <strong>of</strong> <strong>the</strong> lesion consists <strong>of</strong> dentinrecognisable by <strong>the</strong> presence <strong>of</strong> tubuli. Enamel plays aminor role, usually confined to small rims in cavities in<strong>the</strong> dentin mass. The stroma consists <strong>of</strong> mature fibrousconnective tissue.Sometimes, odontomas may contain areas identicalto <strong>the</strong> calcifying odontogenic cyst includingghost cells [22, 61, 80]. Odontoma-like structuresmay also occur in <strong>the</strong> hyperplastic dental follicle (seeSect. 4.4.2.1).Complex odontomas may reveal <strong>the</strong>ir existence bydisturbances in tooth eruption, missing teeth or jawexpansion. Quite <strong>of</strong>ten, <strong>the</strong>y are incidental findings onradiographs taken for o<strong>the</strong>r purposes. In those cases,an amorphous calcified mass is seen that may be connectedwith <strong>the</strong> crown <strong>of</strong> an unerupted tooth. Treatmentconsists <strong>of</strong> conservative removal. Recurrences arenot seen.4.4.3.4 Odontoma –Compound TypeICD-O:9281/0Compound odontoma is a malformation consisting<strong>of</strong> tiny teeth that may vary in number from only a fewto numerous. These teeth do not resemble <strong>the</strong> normal


118 P.J. Slootweg4teeth, but are usually cone-shaped. Histologically, <strong>the</strong>yshow <strong>the</strong> normal arrangement <strong>of</strong> centrally placed fibrovascularpulp tissue surrounded by dentin with an outersurface covered by enamel in <strong>the</strong> crown area <strong>and</strong> cementumin <strong>the</strong> root part.Compound odontoma is one <strong>of</strong> <strong>the</strong> more frequentlyencountered odontogenic lesions [34, 127]. Data on age<strong>of</strong> occurrence show <strong>the</strong> same wide range as with complexodontoma: 0.5–73 years <strong>of</strong> age, which is due to <strong>the</strong>fact that compound odontomas may also remain unnoticedfor a long time.In contrast with almost all o<strong>the</strong>r odontogenic lesionsthat have <strong>the</strong> posterior m<strong>and</strong>ible as <strong>the</strong> preferredsite, compound odontomas have a definite predilectionfor <strong>the</strong> anterior maxilla [118, 153]. They may causeswelling or disturbed tooth eruption. Radiographically,a radiolucency containing multiple tooth-like radiopaquestructures is seen. Treatment consists <strong>of</strong> enucleation<strong>and</strong> <strong>the</strong>re is no recurrence.Compound odontomas may contain areas identicalto those <strong>of</strong> <strong>the</strong> calcifying odontogenic cyst includingghost cells (see Sect. 4.4.3.6) [22, 61, 80].4.4.3.5 Odonto-AmeloblastomaICD-O:9311/0Odonto-ameloblastoma is a very rare neoplasm thatcombines <strong>the</strong> features <strong>of</strong> ameloblastoma <strong>and</strong> odontoma,including <strong>the</strong> presence <strong>of</strong> enamel <strong>and</strong> dentin [73,181]. The ameloblastoma component determines clinicalpresentation <strong>and</strong> behaviour. Radiographically, <strong>the</strong>soap-bubble appearance <strong>of</strong> ameloblastoma is combinedwith radiopaque masses due to <strong>the</strong> odontoma component[97].4.4.3.6 CalcifyingOdontogenic CystFig. 4.24. Calcifying odontogenic cyst closely resembles ameloblastoma,but <strong>the</strong> presence <strong>of</strong> large intraepi<strong>the</strong>lial aggregates <strong>of</strong>ghost cells rules out <strong>the</strong> latter diagnosisICD-O:9301/0Calcifying odontogenic cysts occur both in <strong>the</strong> maxilla<strong>and</strong> in <strong>the</strong> m<strong>and</strong>ible in equal proportions. They mostcommonly lie intraosseously, but cases in <strong>the</strong> gingiva arealso seen. The lesion is most commonly found in <strong>the</strong> 2nd<strong>and</strong> 3rd decades [19, 20, 173].In its most simple form, calcifying odontogenic cystis a cavity with a fibrous wall <strong>and</strong> an epi<strong>the</strong>lial lining.This epi<strong>the</strong>lial lining closely mimics that seen in unicysticameloblastoma, but, in addition, <strong>the</strong>re are intraepi<strong>the</strong>lialeosinophilic ghost cells lacking nuclei that mayundergo calcification. Ghost cell masses may also herniatethrough <strong>the</strong> basal lamina to reach <strong>the</strong> underlyingstroma where <strong>the</strong>y can act as foreign material <strong>and</strong> evokea giant cell reaction (Fig. 4.24). In <strong>the</strong> fibrous stroma adjacentto <strong>the</strong> basal epi<strong>the</strong>lial cells, homogenous eosinophilicmaterial resembling dentin may be found in varyingamounts. Dentin-like material <strong>and</strong> ghost cells toge<strong>the</strong>rmay form mixed aggregates.To this simple unicystic structure o<strong>the</strong>r features maybe added, thus creating different subtypes with differentnames [173].The proliferative calcifying odontogenic cyst showsmultiple intramural daughter cysts with an epi<strong>the</strong>liallining similar to <strong>the</strong> main cyst cavity. The solid (neoplastic)calcifying odontogenic cyst has been describedby a variety <strong>of</strong> o<strong>the</strong>r terms: dentinogenic ghost cell tumour,epi<strong>the</strong>lial odontogenic ghost cell tumour, calcifyingghost cell odontogenic tumour, <strong>and</strong> cystic calcifyingodontogenic tumour [81]. This lesion combines <strong>the</strong>morphology <strong>of</strong> an ameloblastoma with intra-epi<strong>the</strong>lial<strong>and</strong> stromal ghost cells with a dentin-like material.The most recent WHO classification proposes <strong>the</strong>diagnostic designations calcifying cystic odontogenictumour (ICD-O:9301/0) <strong>and</strong> dentinogenic ghost celltumour (ICD-O:9302/0) to discern between <strong>the</strong> cystic<strong>and</strong> <strong>the</strong> solid lesion [181].Also, calcifying odontogenic cysts may occur in associationwith o<strong>the</strong>r odontogenic tumours, in most instancesameloblastoma <strong>and</strong> odontoma [124]. All variants<strong>of</strong> calcifying odontogenic cyst may show melaninpigment [60, 162].Ghost cells, ei<strong>the</strong>r intraepi<strong>the</strong>lially or in <strong>the</strong> stroma,separate calcifying odontogenic cyst from ameloblastoma.The solid variant <strong>of</strong> calcifying odontogenic cyst issimilar to craniopharyngioma [7, 111].Intraosseous calcifying odontogenic cysts cause bonyexpansion. The peripheral type forms a gingival s<strong>of</strong>t tissueswelling. Radiographically, <strong>the</strong> lesion shows a lucentappearance when located intraosseously, mostly withvariable amounts <strong>of</strong> radiopacities. Treatment <strong>of</strong> <strong>the</strong> calcifyingodontogenic cyst consists <strong>of</strong> enucleation in cases<strong>of</strong> intraosseous location or excision for peripheral ones.


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 119Recurrences have been documented for both <strong>the</strong> cystic<strong>and</strong> <strong>the</strong> solid variant [124, 143]. If <strong>the</strong> lesion is combinedwith an ameloblastoma, this latter component dictates<strong>the</strong> most appropriate <strong>the</strong>rapy, which consists <strong>of</strong> surgicalexcision including a margin <strong>of</strong> healthy tissue as alreadydiscussed [124].4.4.4 Odontogenic Tumours –MalignantBoth odontogenic epi<strong>the</strong>lium as well as odontogenicmesenchyme may show neoplastic degeneration, causingei<strong>the</strong>r odontogenic carcinomas or odontogenicsarcomas [73, 159, 181]. All entities to be mentionedshow <strong>the</strong> clinical presentation <strong>and</strong> course as well as <strong>the</strong>radiographic appearance <strong>of</strong> an intraosseous malignanttumour.4.4.4.1 Malignant AmeloblastomaICD-O:9310/3Malignant (metastasising) ameloblastoma is an ameloblastomathat metastasises in spite <strong>of</strong> an innocuoushistologic appearance. The primary tumour shows nospecific features that are different from ameloblastomasthat do not metastasise. Therefore, this diagnosiscan only be made in retrospect, after <strong>the</strong> occurrence <strong>of</strong>metastatic deposits. It is thus clinical behaviour <strong>and</strong> nothistology that justifies a diagnosis <strong>of</strong> malignant ameloblastoma[181]. This definition pr<strong>of</strong>oundly differs fromthat given in <strong>the</strong> previous WHO classification [73]. Atthat time a malignant ameloblastoma was described asa neoplasm in which <strong>the</strong> pattern <strong>of</strong> an ameloblastomais combined with cytological features <strong>of</strong> malignancy,a definition not based on behaviour but on histology.It is obvious that disparate views on what representsa malignant ameloblastoma can give rise to confusion[41, 159]. It has to be emphasised that to avoid mixingup different entities, <strong>the</strong> term malignant ameloblastomashould be reserved for metastasising ameloblastomaswhereas <strong>the</strong> ameloblastomas with atypia shouldbe called ameloblastic carcinomas, a type <strong>of</strong> lesion tobe discussed in Sect. 4.4.4.2 [104]. Confusion may alsoarise through <strong>the</strong> use <strong>of</strong> <strong>the</strong> term atypical ameloblastomasto denote lesions with a fatal outcome for variousreasons, ei<strong>the</strong>r metastasis, histologic atypia or relentlesslocal spread [3].Metastatic deposits <strong>of</strong> malignant ameloblastomas aremostly seen in <strong>the</strong> lung [76, 77, 159]. Apart from metastasis,malignant ameloblastoma shows no featuresthat are different from conventional ameloblastoma (seeSect. 4.4.1.1).Fig. 4.25. Ameloblastic carcinoma combines <strong>the</strong> presence <strong>of</strong> epi<strong>the</strong>lialnests with peripheral palisading <strong>and</strong> cytonuclear atypia4.4.4.2 Ameloblastic CarcinomaICD-O:9270/3Ameloblastic carcinoma , an entity that has only recentlybeen recognised, is a lesion with <strong>the</strong> histologicfeatures <strong>of</strong> both ameloblastoma <strong>and</strong> squamous cellcarcinoma [42, 181]. This tumour may arise de novoor from a pre-existing benign odontogenic tumour orcyst [159].Most cases <strong>of</strong> ameloblastic carcinoma occur in <strong>the</strong>m<strong>and</strong>ible [30]. They show a wide age range with a mean<strong>of</strong> 30.1 years [30].Ameloblastic carcinoma is characterised by cells that,although mimicking <strong>the</strong> architectural pattern <strong>of</strong> ameloblastoma,exhibit pronounced cytological atypia <strong>and</strong>mitotic activity, thus allowing <strong>the</strong> distinction betweenameloblastic carcinoma <strong>and</strong> ameloblastoma (Fig. 4.25).Metastatic lesions are described in <strong>the</strong> lungs <strong>and</strong> in <strong>the</strong>lymph nodes [36, 151].4.4.4.3 PrimaryIntraosseous CarcinomaICD-O:9270/3Primary intraosseous carcinoma is a squamous cell carcinomaarising within <strong>the</strong> jaw, having no initial connectionwith <strong>the</strong> oral mucosa, <strong>and</strong> presumably developingfrom residues <strong>of</strong> <strong>the</strong> odontogenic epi<strong>the</strong>lium [73, 181].It ranges from well to poorly differentiated [42], mainlyoccurs in <strong>the</strong> posterior m<strong>and</strong>ible <strong>and</strong> is more <strong>of</strong>ten seenin males [190].The tumour may arise from a still recognisable precursorlesion such as <strong>the</strong> epi<strong>the</strong>lial lining <strong>of</strong> an odontogeniccyst [67, 90]. Also, enamel epi<strong>the</strong>lium has beendocumented as a tissue <strong>of</strong> origin [154].


120 P.J. Slootweg4Fig. 4.26. Clear cell odontogenic carcinoma is characterised byclear cells forming epi<strong>the</strong>lial nestsFig. 4.27. At higher magnification, clear cell odontogenic carcinomais seen to contain clear cells as well as eosinophilic cellsSwelling <strong>of</strong> <strong>the</strong> jaw <strong>and</strong> pain are most <strong>of</strong>ten <strong>the</strong> presentingsigns. Surgery <strong>and</strong> postoperative radio<strong>the</strong>rapyseem to provide <strong>the</strong> best results. The tumour maymetastasise to regional lymph nodes as well as lungs.Prognosis is poor with almost 50% <strong>of</strong> <strong>the</strong> patients failingloco-regionally within <strong>the</strong> first 2 years <strong>of</strong> followup[190].4.4.4.4 Clear CellOdontogenic CarcinomaICD-O:9341/1Clear cell odontogenic carcinoma was initially reportedas a clear cell odontogenic tumour [56]. As <strong>the</strong>se lesionsnot only behave aggressively locally, but may also metastasise,<strong>the</strong> currently used diagnostic term clear cellodontogenic carcinoma appears to be more appropriate[43, 75]. The tumour is mostly seen in elderly patients[5].The tumour is composed <strong>of</strong> cells with clear cytoplasm.These cells form nests <strong>and</strong> str<strong>and</strong>s, intermingledwith smaller isl<strong>and</strong>s <strong>of</strong> cells with eosinophilic cytoplasm(Figs. 4.26, 4.27). Also, squamous differentiationhas been reported [17]. Cells at <strong>the</strong> periphery <strong>of</strong><strong>the</strong> nests may show palisading. The clear cells stain positivefor glycogen as well as for epi<strong>the</strong>lial markers keratinAE1/AE3, cytokeratin 8/18, cytokeratin 19 <strong>and</strong> epi<strong>the</strong>lialmembrane antigen (EMA) [89].The anterior m<strong>and</strong>ible appears to be <strong>the</strong> site <strong>of</strong> predilection.Metastases are found in lymph nodes as well asin <strong>the</strong> lungs <strong>and</strong> <strong>the</strong> skeleton. Recurrent disease is seenin more than 50% <strong>of</strong> cases with documented follow-up.Death due to <strong>the</strong> tumour has also been reported [5, 89].Differential diagnosis includes metastatic renal cellcarcinoma, <strong>the</strong> clear cell variant <strong>of</strong> mucoepidermoidcarcinoma, <strong>and</strong> ameloblastoma with clear cells. Metastaticrenal cell carcinoma may be ruled out on clinicalgrounds. The clear cell variant <strong>of</strong> muco-epidermoidcarcinoma can be identified with stains for mucin production.Differentiation from ameloblastoma with clearcells may be problematic <strong>and</strong> it has been proposed that<strong>the</strong>se lesions represent <strong>the</strong> same entity [16]. Clear cellcarcinoma <strong>of</strong> minor salivary gl<strong>and</strong> origin is ano<strong>the</strong>r differentialdiagnosis (see Chap. 5).4.4.4.5 Malignant Epi<strong>the</strong>lialOdontogenicGhost Cell TumourICD-O:9270/3Malignant epi<strong>the</strong>lial odontogenic ghost cell tumour ,also called odontogenic ghost cell carcinoma is a tumourthat combines <strong>the</strong> elements <strong>of</strong> a benign calcifying odontogeniccyst with a malignant epi<strong>the</strong>lial component.Only a few cases <strong>of</strong> this tumour have been reported, thusprecluding any conclusions regarding clinicopathologicfeatures. Malignancy has been demonstrated by local aggressivegrowth <strong>and</strong> distant metastasis [84]. The tumourapparently arises most <strong>of</strong>ten from malignant transformation<strong>of</strong> a pre-existing benign calcifying odontogeniccyst [81].4.4.4.6 Odontogenic SarcomaICD-O:9270/3The WHO discerns between ameloblastic fibrosarcoma ,ameloblastic fibrodentino- <strong>and</strong> fibro-odontosarcoma<strong>and</strong> odontogenic carcinosarcoma [73, 181]. The ameloblasticfibrosarcoma consists <strong>of</strong> malignant connectivetissue admixed with epi<strong>the</strong>lium similar to that seen inan ameloblastoma or ameloblastic fibroma [152]. If <strong>the</strong>reis also dentin, this is known as an ameloblastic fibrodentinosarcoma, <strong>and</strong> if <strong>the</strong>re is also enamel, it is called ameloblasticfibro-odontosarcoma . This subclassification hasno prognostic significance [2]. These tumours may arise


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 121Table 4.3. Fibro-osseous lesions [13, 181]Fibrous dysplasiaOssifying fibromaOsseous dysplasiaConventionalJuvenile trabecularJuvenile psammomatoidPeriapical osseous dysplasiaFocal osseous dysplasiaFlorid osseous dysplasiaFamilial gigantiform cementomade novo or from a pre-existing ameloblastic fibroma orameloblastic fibro-odontoma [98].Those extremely rare lesions that combine carcinomatous<strong>and</strong> sarcomatous elements, but are recognisableas odontogenic from <strong>the</strong> epi<strong>the</strong>lial component, resembleameloblastic carcinomas, <strong>and</strong> have been called odontogeniccarcinosarcoma or odontogenic carcinoma with sarcomatousproliferation [152].4.5 Fibro-Osseous LesionsThe current classification <strong>of</strong> maxill<strong>of</strong>acial fibro-osseouslesions includes fibrous dysplasia, ossifying fibroma <strong>and</strong>osseous dysplasia [13, 158]. Table 4.3 gives an overview<strong>of</strong> <strong>the</strong> various entities in this group.4.5.1 Fibrous DysplasiaFibrous dysplasia is composed <strong>of</strong> cellular fibrous tissuecontaining trabeculae <strong>of</strong> woven bone. It occurs in threeclinical subtypes: monostotic, which affects one bone,polyostotic, which affects multiple bones, <strong>and</strong> Albright’ssyndrome, in which multiple bone lesions are accompaniedby skin hyperpigmentation <strong>and</strong> endocrine disturbances[13]. Activating missense mutations <strong>of</strong> <strong>the</strong> geneencoding <strong>the</strong> α subunit <strong>of</strong> <strong>the</strong> stimulatory G protein area consistent finding in <strong>the</strong> various forms <strong>of</strong> fibrous dysplasia[123].Crani<strong>of</strong>acial fibrous dysplasia is usually <strong>of</strong> <strong>the</strong> monostotictype [44]. The disease mostly occurs during <strong>the</strong>first three decades, although cases are occasionally seenat an older age. The maxilla is more <strong>of</strong>ten involved than<strong>the</strong> m<strong>and</strong>ible. In <strong>the</strong> maxilla, fibrous dysplasia may extendby continuity across suture lines to involve adjacentbones [13].Fibrous dysplasia shows replacement <strong>of</strong> <strong>the</strong> normalbone by moderately cellular fibrous tissue containingirregularly shaped trabeculae consisting <strong>of</strong> wovenbone without rimming osteoblasts that fuse with adjacentbone. Jaw lesions may also show lamellar bone(Fig. 4.28). Sometimes, tiny calcified spherules may bepresent [158].Fig. 4.28. In <strong>the</strong> jaws, fibrous dysplasia may consist <strong>of</strong> both woven<strong>and</strong> lamellar bone, as shown in this photomicrograph takenwith <strong>the</strong> use <strong>of</strong> partly polarised light to enhance <strong>the</strong> collagenousscaffold <strong>of</strong> <strong>the</strong> boneFibrous dysplasia has to be distinguished from o<strong>the</strong>rlesions characterised by <strong>the</strong> combination <strong>of</strong> fibrous tissue<strong>and</strong> bone: ossifying fibroma, osseous dysplasia, lowgradeosteosarcoma <strong>and</strong> sclerosing osteomyelitis. However,none <strong>of</strong> <strong>the</strong>se is composed <strong>of</strong> woven bone trabeculaefusing with adjacent uninvolved bone. Ossifying fibroma<strong>and</strong> osseous dysplasia both show much variety inappearance <strong>of</strong> mineralised material <strong>and</strong> stromal cellularity,low-grade osteosarcoma invades through <strong>the</strong> corticalbone into s<strong>of</strong>t tissues <strong>and</strong> sclerosing osteomyelitisshows coarse trabeculae <strong>of</strong> lamellar bone, whereas <strong>the</strong>intervening stroma is not cellular but oedematous withsprinkled lymphocytes [158].Fibrous dysplasia clinically presents as a painlessswelling <strong>of</strong> <strong>the</strong> bone involved. Radiographically, <strong>the</strong> classicalappearance is described as orange-skin or groundglassradiopacity without defined borders [13]. Usually,fibrous dysplasia is a self-limiting disease. Therefore,treatment is only required if <strong>the</strong>re are problems due tolocal increase in size <strong>of</strong> <strong>the</strong> affected bone. Sometimes, anosteosarcoma may arise in fibrous dysplasia [39].4.5.2 Ossifying FibromaICD-O:9262/0, 9274/0Ossifying fibroma , formerly also called cemento-ossifyingfibroma is a well-demarcated lesion composed <strong>of</strong>fibrocellular tissue <strong>and</strong> mineralised material <strong>of</strong> varyingappearance. It occurs most <strong>of</strong>ten in <strong>the</strong> 2nd through <strong>the</strong>4th decades. The lesion shows a predilection for females,is mostly seen in <strong>the</strong> posterior m<strong>and</strong>ible [13] <strong>and</strong> mayoccur multifocally [70].Chromosomal abnormalities have been observed inossifying fibromas [31, 49, 138]. Data are still too scarceto determine <strong>the</strong>ir pathogenetic significance.


122 P.J. Slootweg4Fig. 4.29. Ossifying fibroma contains both cell-rich <strong>and</strong> cell-poorareas as well as well-structured bone <strong>and</strong> amorphous calcified materialFig. 4.31. Juvenile trabecular ossifying fibroma shows slenderbony trabeculae rimmed with osteoblasts that merge with an extremelycellular stromaFig. 4.30. Ossifying fibroma may also contain more smoothlycontoured bony elements, formerly thought to represent cementumFig. 4.32. At higher magnification, <strong>the</strong> plump osteoblasts thatline <strong>the</strong> bony trabeculae in juvenile trabecular ossifying fibromaare shown to be a prominent featureOssifying fibroma is composed <strong>of</strong> fibrous tissue thatmay vary in cellularity from areas with closely packedcells displaying mitotic figures to almost acellular sclerosingparts within one <strong>and</strong> <strong>the</strong> same lesion. The mineralisedcomponent may consist <strong>of</strong> plexiform bone, lamellar bone<strong>and</strong> acellular mineralised material, sometimes all occurringtoge<strong>the</strong>r in one single lesion (Figs 4.29, 4.30).Juvenile psammomatoid <strong>and</strong> juvenile trabecular ossifyingfibroma are subtypes [40]. Juvenile trabecularossifying fibroma consists <strong>of</strong> cell-rich fibrous tissuewith b<strong>and</strong>s <strong>of</strong> cellular osteoid toge<strong>the</strong>r with slendertrabeculae <strong>of</strong> plexiform bone lined by a dense rim<strong>of</strong> enlarged osteoblasts (Figs. 4.31, 4.32). Sometimes<strong>the</strong>se trabeculae may anastomose to form a lattice.Mitoses are present, especially in <strong>the</strong> cell-rich areas.Also, multinucleated giant cells, pseudocystic stromaldegeneration <strong>and</strong> haemorrhages may be present. Dueto its cellularity <strong>and</strong> mitotic activity, <strong>the</strong> lesion may beconfused with osteosarcoma. However, atypical cellularfeatures or abnormal mitotic figures are not seen.Moreover, <strong>the</strong> lesion is demarcated from its surroundings[73, 158, 181].Juvenile psammomatoid ossifying fibroma is characterisedby a fibroblastic stroma containing small ossiclesresembling psammoma bodies, hence its name. Thestroma varies from loose <strong>and</strong> fibroblastic to intenselycellular. The spherical or curved ossicles are acellularor include sparsely distributed cells (Fig. 4.33). Theyshould not be confused with <strong>the</strong> cementum-like depositsthat are present in conventional ossifying fibroma.These particles have a smooth contour whereas <strong>the</strong> ossiclesin juvenile psammomatoid ossifying fibroma hasa peripheral radiating fringe <strong>of</strong> collagen fibres. Ossiclesmay coalesce to form trabeculae. Sometimes, juvenilepsammomatoid ossifying fibroma contains basophilic,concentrically lamellated particles, as well as irregularthread-like or thorn-like calcified str<strong>and</strong>s in a hyalinisedbackground (Fig. 4.34). O<strong>the</strong>r features such as trabeculae<strong>of</strong> woven bone as well as lamellar bone, pseudocysticstromal degeneration <strong>and</strong> haemorrhages resulting


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 123Fig. 4.33. Psammomatoid ossicles in a cellular stroma characterisejuvenile psammomatoid ossifying fibroma. A comparison withFig. 4.30 shows that <strong>the</strong>se particles are not as smoothly outlined asthose occurring in conventional ossifying fibromasFig. 4.35. Osseous dysplasia typically lies at <strong>the</strong> root tip. It consists<strong>of</strong> fibrous tissue containing mineralised material <strong>of</strong> varyingappearanceFig. 4.34. Thread-like calcifications in an eosinophilic matrix arealso <strong>of</strong>ten present in juvenile psammomatoid ossifying fibroma<strong>the</strong> psammomatoid ossicles in juvenile psammomatoidossifying fibroma are clearly different from <strong>the</strong> acellularspherical real psammoma bodies [172].Clinically, ossifying fibroma causes expansion <strong>of</strong> <strong>the</strong>bone involved leading to a palpable swelling. Radiographically,a demarcated lesion is seen that may haveradiodense as well as radiolucent areas depending on<strong>the</strong> various contributions <strong>of</strong> s<strong>of</strong>t <strong>and</strong> hard tissue componentsto an individual lesion [13].Excision <strong>of</strong> ossifying fibromas usually yields a permanentcure.in areas similar to an aneurysmal bone cyst, multinucleategiant cells, <strong>and</strong> mitotic figures can also be observed.Juvenile psammomatoid ossifying fibroma has<strong>the</strong> bony walls <strong>of</strong> <strong>the</strong> paranasal sinuses as site <strong>of</strong> predilection[179].Ossifying fibroma may be confused with fibrous dysplasia.The most important distinguishing feature is <strong>the</strong>presence <strong>of</strong> demarcation <strong>and</strong>/or encapsulation in ossifyingfibroma as opposed to <strong>the</strong> merging with its surroundingsas shown by fibrous dysplasia. In addition,<strong>the</strong> variation in cellularity as well as in appearance <strong>of</strong>mineralised material distinguishes ossifying fibromafrom fibrous dysplasia. To distinguish ossifying fibromafrom osseous dysplasia, data on clinical presentation<strong>and</strong> radiographic appearance are indispensable (seeSect. 4.5.3).Juvenile psammomatoid ossifying fibroma has to bedifferentiated from meningioma with psammoma bodies;immunohistochemistry positive for EMA rules outjuvenile psammomatoid ossifying fibroma. Moreover,4.5.3. Osseous DysplasiaICD-O:9272/0Osseous dysplasia is a pathologic process <strong>of</strong> unknownaetiology located in <strong>the</strong> tooth-bearing jaw areas in <strong>the</strong>vicinity <strong>of</strong> <strong>the</strong> tooth apices <strong>and</strong> is thought to arise from<strong>the</strong> proliferation <strong>of</strong> periodontal ligament fibroblaststhat may deposit bone as well as cementum. The conditionoccurs in various clinical forms that bear differentnames. However, all have <strong>the</strong> same histomorphology:cellular fibrous tissue, trabeculae <strong>of</strong> woven as well aslamellar bone <strong>and</strong> spherules <strong>of</strong> cementum-like material(Fig. 4.35). The ratio <strong>of</strong> fibrous tissue to mineralisedmaterial may vary <strong>and</strong> it has been shown that <strong>the</strong>se lesionsare initially fibroblastic, but over <strong>the</strong> course <strong>of</strong>several years may show increasing degrees <strong>of</strong> calcification.This variation in ratio <strong>of</strong> s<strong>of</strong>t tissue to hard tissueis reflected in <strong>the</strong> radiographic appearance; lesions arepredominantly radiolucent, predominantly radiodenseor mixed.


124 P.J. Slootweg4Fig. 4.36. Central giant cell granuloma shows osteoclast-like giantcells in a loose fibrocellular stroma. Cherubism has an identicalappearanceOsseous dysplasia lacks encapsulation or demarcation,but tends to merge with <strong>the</strong> adjacent cortical ormedullary bone [13].The several subtypes <strong>of</strong> osseous dysplasia are distinguishedby clinical <strong>and</strong> radiological features. Periapicalosseous dysplasia occurs in <strong>the</strong> anterior m<strong>and</strong>ible <strong>and</strong>involves only a few adjacent teeth. A similar limited lesionoccurring in a posterior jaw quadrant is known asfocal osseous dysplasia [167]. Florid osseous dysplasia isnon-expansile, involves two or more jaw quadrants <strong>and</strong>occurs in middle-aged black females [13]. Familial gigantiformcementoma is expansile, involves multiplequadrants <strong>and</strong> occurs at a young age. This type <strong>of</strong> osseousdysplasia shows an autosomal dominant inheritancewith variable expression, but sporadic cases without ahistory <strong>of</strong> familial involvement have also been reported[1, 189]. Simple bone cysts may be seen with florid <strong>and</strong>focal osseous dysplasia [59, 62].Osseous dysplasia has to be distinguished from ossifyingfibroma. Osseous dysplasia is a mixed radiolucent-radiodenselesion with ill-defined borders in <strong>the</strong>tooth-bearing part <strong>of</strong> <strong>the</strong> jaws, ei<strong>the</strong>r localised or occupyinglarge jaw areas depending on <strong>the</strong> type. In contrast,ossifying fibroma is usually a localised lesion thatexp<strong>and</strong>s <strong>the</strong> jaw, <strong>and</strong> is predominantly radiolucent withradiodense areas [164].Osseous dysplasia also has to be differentiated fromsclerosing osteomyelitis [140]. Sclerotic lamellar bonetrabeculae <strong>and</strong> well-vascularised fibrous tissue withlymphocytes <strong>and</strong> plasma cells define sclerosing osteomyelitis,whereas cementum-like areas <strong>and</strong> fibrocellulars<strong>of</strong>t tissue are lacking [53].The various forms <strong>of</strong> osseous dysplasia do not requiretreatment unless necessitated by complications such asinfection <strong>of</strong> sclerotic bone masses, as may occur in floridosseous dysplasia, or facial deformity, as may be seenin familial gigantiform cementoma.4.6 Giant Cell LesionsCentral giant cell granuloma , cherubism <strong>and</strong> aneurysmalbone cyst all show osteoclast-like giant cells lyingin a fibroblastic background tissue. The fibroblastic tissuemay vary in cellularity from very dense to cell-poor.Mitotic figures may be encountered but are usually notnumerous <strong>and</strong> not atypical. The giant cells mostly clusterin areas <strong>of</strong> haemorrhage, but <strong>the</strong>y may also lie moredispersed among <strong>the</strong> lesion (Fig. 4.36). Bone formation,if present, is usually confined to <strong>the</strong> periphery <strong>of</strong> <strong>the</strong> lesion.Radiologically, all three types <strong>of</strong> giant cell lesionshave a lucent, quite <strong>of</strong>ten multilocular appearance. Multiplegiant cell lesions may occur in association withNoonan’s syndrome as well as with neur<strong>of</strong>ibromatosis[38, 134]. Fur<strong>the</strong>r discussion will only include giant cellgranuloma <strong>and</strong> cherubism as <strong>the</strong>y are confined to <strong>the</strong>jaws.4.6.1 Central Giant Cell GranulomaCentral giant cell granuloma is mostly seen before <strong>the</strong>age <strong>of</strong> 30. The lesion is restricted to <strong>the</strong> jaws, <strong>the</strong> m<strong>and</strong>iblebeing more <strong>of</strong>ten involved than <strong>the</strong> maxilla. Its aetiologyis unknown. Lesions with a histologic appearanceidentical to that <strong>of</strong> <strong>the</strong> central giant cell granuloma mayoccur in <strong>the</strong> gingiva <strong>and</strong> are called giant cell epulis (seeChap. 3). Sometimes, lesions combine <strong>the</strong> appearance<strong>of</strong> a giant cell granuloma with that <strong>of</strong> an odontogenicfibroma [108].Clinically, central giant cell granuloma manifests itselfas a localised jaw swelling. Radiographically, it is aradiolucent lesion that may be ei<strong>the</strong>r uni- or multilocular.As <strong>the</strong> lesion is not encapsulated, removal is sometimesfollowed by recurrence. If <strong>the</strong>re is recurrence, hyperparathyroidismshould be ruled out as <strong>the</strong> brown tumoursassociated with this latter disease are identical togiant cell granulomas. Also, distinction has to be madebetween giant cell granulomas <strong>and</strong> true giant cell tumours.There has been much discussion whe<strong>the</strong>r giantcell granulomas with more aggressive behaviour thanusually observed could represent a gnathic manifestation<strong>of</strong> this latter lesion [4, 163].4.6.2 CherubismIn cases <strong>of</strong> cherubism, two or more jaw quadrants containlesions histologically similar to giant cell granuloma.The disease occurs in young children, <strong>of</strong>ten with ahistory <strong>of</strong> o<strong>the</strong>r afflicted family members. The geneticdefect responsible for cherubism has been localised tochromosome 4p16.3 [112].The expansion <strong>of</strong> <strong>the</strong> affected jaw areas causes <strong>the</strong> angelicface leading to <strong>the</strong> lesion’s designation: cherubism.


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 125With <strong>the</strong> onset <strong>of</strong> puberty, <strong>the</strong> lesions lose <strong>the</strong>ir activity<strong>and</strong> may mature to fibrous tissue <strong>and</strong> bone.There may also be a component consisting <strong>of</strong> immatureodontogenic tissue due to developing tooth germslying within <strong>the</strong> lesional tissue. This is a fortuitous findingwithout any clinical relevance.4.7 Neoplastic Lesions<strong>of</strong> <strong>the</strong> Maxill<strong>of</strong>acial Bones,Non-OdontogenicTo be included in <strong>the</strong> following text, lesions should bemainly confined to <strong>the</strong> maxill<strong>of</strong>acial bones.Fig. 4.37. At high magnification, <strong>the</strong> vacuolated nature <strong>of</strong> <strong>the</strong>chordoma cells are clearly visible, as is <strong>the</strong>ir epi<strong>the</strong>lial cohesion4.7.1 OsteomaICD-O:9180/0Osteomas are lesions composed <strong>of</strong> compact lamellarbone with sparse marrow cavities filled with fatty orfibrous tissue. In <strong>the</strong> maxill<strong>of</strong>acial skeleton, <strong>the</strong>y mostcommonly occur in <strong>the</strong> frontal <strong>and</strong> ethmoid sinus; less<strong>of</strong>ten, <strong>the</strong> maxillary antrum <strong>and</strong> <strong>the</strong> sphenoid sinus areinvolved [137]. They may also occur in <strong>the</strong> jaw bones asa manifestation <strong>of</strong> Gardner’s syndrome [183].Paranasal osteomas as a group are common lesions[92]. Clinically, <strong>the</strong>y cause sinusitis <strong>and</strong> headache or o<strong>the</strong>rsigns <strong>of</strong> sinonasal disease. Similar bony outgrowths at<strong>the</strong> palate or m<strong>and</strong>ible are called tori.4.7.2 ChordomaICD-O:9370/3Chordomas are malignant tumours derived from embryonicremnants <strong>of</strong> <strong>the</strong> notochord. The mostly occur atei<strong>the</strong>r <strong>the</strong> cranial or caudal end <strong>of</strong> <strong>the</strong> vertebral column[57]. Chordomas show a slight male predominance; <strong>the</strong>ymay occur at any age [57].Three different types <strong>of</strong> chordoma are discerned:conventional, chondroid, <strong>and</strong> dedifferentiated. Conventionalchordoma consists <strong>of</strong> lobules separated fromeach o<strong>the</strong>r by fibrous b<strong>and</strong>s. These lobules containovoid cells with small, dark nuclei <strong>and</strong> homogeneouseosinophilic cytoplasm. O<strong>the</strong>r cells show large vesicularnuclei <strong>and</strong> abundant cytoplasm with vacuoles.Sometimes, <strong>the</strong>se cells contain only one single vacuolecausing a signet-ring appearance or vacuoles surrounding<strong>the</strong> nucleus: <strong>the</strong>se latter cells represent <strong>the</strong>so-called physaliphorous cells thought to be pathognomonicfor chordoma. In general, <strong>the</strong> cell density ismaximal at <strong>the</strong> periphery <strong>of</strong> <strong>the</strong> lobules; more centrally,<strong>the</strong> cells lose <strong>the</strong>ir epi<strong>the</strong>lial cohesion <strong>and</strong> may lieisolated in an abundant mucoid matrix (Fig. 4.37). Although<strong>the</strong>re may be atypia, mitotic figures are infrequent.The lesion invades adjacent structures. Immunohistochemically,chordoma is characterised by positivityfor S-100 as well as vimentin <strong>and</strong> a broad variety<strong>of</strong> epi<strong>the</strong>lial markers [131].Chondroid chordoma (ICD-O:9371/3) denotes a variant<strong>of</strong> chordoma that contains cartilaginous areas indistinguishablefrom chondrosarcoma [57]. However,chondrocytic differentiation in chordomas probablyrepresents a focal maturation process [51]. Nei<strong>the</strong>rhas <strong>the</strong> distinction between conventional <strong>and</strong> chondroidchordoma any clinical significance [29]. Therefore, it isadvocated that this designation be dropped. Dedifferentiatedchordomas are those lesions that contain areas <strong>of</strong>chordoma as well as an additional malignant mesenchymalcomponent that may be a fibrosarcoma, an osteosarcomaor, most likely, a poorly differentiated sarcoma[64].Chordoma has to be distinguished from chondrosarcoma.Positivity for epi<strong>the</strong>lial markers is a consistentfeature in chordomas <strong>and</strong> is absent in chondrosarcoma[132]. O<strong>the</strong>r look-alikes, such as extraskeletal myxoidchondrosarcoma, myxoid liposarcoma <strong>and</strong> myxopapillaryependymoma, also lack positivity for epi<strong>the</strong>lialmarkers [28]. Chordoid meningiomas may also mimicchordomas, but <strong>the</strong>re are no physaliphorous cells, noris <strong>the</strong>re positivity for cytokeratins in this meningiomasubtype [125].The differential diagnosis <strong>of</strong> chordoma should alsoinclude pleomorphic adenoma. Both lesions may showepi<strong>the</strong>lial clusters as well as single cells with vacuolatedcytoplasm lying in a mucoid matrix. Moreover, positivityfor S-100, vimentin, <strong>and</strong> epi<strong>the</strong>lial markers is displayedby both. Positivity for myoepi<strong>the</strong>lial markers,however, is restricted to pleomorphic adenoma.Chordomas manifest by destroying adjacent structuresresulting in cranial nerve dysfunction. Rarely, <strong>the</strong>ycause a swelling in <strong>the</strong> neck due to lateral growth. Theirsite precludes radical surgical treatment. Mostly, <strong>the</strong>ra-


126 P.J. Slootweg4Fig. 4.38. Melanotic neuroectodermal tumour <strong>of</strong> infancy consists<strong>of</strong> small dark cells <strong>and</strong> larger cells with vesicular nuclei. Melaninis usually associated with <strong>the</strong> latter cell populationpy consists <strong>of</strong> debulking <strong>and</strong> irradiation. Five-year survivalrate is approximately 50%. Histologic features relatedwith prognosis have not been identified [29]. Metastaticdisease is unusual [25].4.7.3 Melanotic NeuroectodermalTumour <strong>of</strong> InfancyICD-O:9363/0Cells derived from <strong>the</strong> neural crest play a major role in<strong>the</strong> formation <strong>of</strong> <strong>the</strong> jaws <strong>and</strong> teeth. These cells are alsothought to be <strong>the</strong> source from which <strong>the</strong> melanotic neuroectodermaltumour <strong>of</strong> infancy develops [106]. Most <strong>of</strong><strong>the</strong> lesions occur before <strong>the</strong> age <strong>of</strong> 1 year. The majority<strong>of</strong> <strong>the</strong>m occur in <strong>the</strong> anterior maxilla [68].The tumour shows dense fibrous stroma with nestscomposed <strong>of</strong> two different cell types: centrally placedsmall dark cells without any discernable cytoplasm <strong>and</strong>peripherally located larger cells with vesicular nuclei<strong>and</strong> ample cytoplasm with melanin pigment (Fig. 4.38)[9, 68, 113]. Maturation <strong>of</strong> <strong>the</strong> small cells to ganglioncells has been reported [144]. Although <strong>the</strong> cells may beatypical, mitotic figures are rare [9]. Sometimes, a transition<strong>of</strong> <strong>the</strong> large cells to osteoblasts forming tiny bonytrabeculae can be observed [157]. The lesion is not encapsulated.Immunohistochemically, <strong>the</strong> large cells are positivefor a wide variety <strong>of</strong> cytokeratins, neuron-specific enolase,S-100, HMB45 <strong>and</strong> chromogranin. The small cellsshow positivity for CD56, neuron-specific enolase, synaptophysin<strong>and</strong> chromogranin [9]. This pattern can besummarised as evidence for neural, melanocytic <strong>and</strong> epi<strong>the</strong>lialdifferentiation. In addition, <strong>the</strong> large cells havebeen shown to be positive for vimentin [157]. Ultrastructurally,<strong>the</strong> small cells show neurosecretory granules<strong>and</strong> <strong>the</strong> large cells show melanosomes at differentstages <strong>of</strong> development [113].Quite <strong>of</strong>ten, immature odontogenic tissues form part<strong>of</strong> <strong>the</strong> material excised or biopsied, due to <strong>the</strong> early age<strong>of</strong> occurrence <strong>and</strong> <strong>the</strong> close association <strong>of</strong> <strong>the</strong> tumourwith tooth germs. This should not be mistaken as evidence<strong>of</strong> an odontogenic tumour. The highly characteristichistological pattern leaves no room for o<strong>the</strong>r differentialdiagnostic considerations.Clinically, melanotic neuroectodermal tumour <strong>of</strong> infancymanifests as a rapidly growing blue tissue mass,usually at <strong>the</strong> anterior alveolar maxillary ridge. Radiologically,bone resorption may be seen, although this isdifficult to evaluate in <strong>the</strong> delicate bony structures <strong>of</strong> <strong>the</strong>infantile maxilla. Tooth germs are displaced <strong>and</strong> may liewithin <strong>the</strong> tumour mass. Conservative excision usuallyconstitutes adequate treatment. Recurrences have beendescribed, but metastases are exceptionally rare [113].There are no histological features predicting more aggressivebehaviour [9].References1. Abdelsayed RA, Eversole LR, Singh BS, Scarbrough FE (2001)Gigantiform cementoma: clinicopathologic presentation <strong>of</strong> 3cases. Oral Surg Oral Med Oral Pathol Oral Radiol Endod91:438–4442. Altini M, Thompson SH, Lownie JF, Berezowski BB (1985)Ameloblastic sarcoma <strong>of</strong> <strong>the</strong> m<strong>and</strong>ible. J Oral Maxill<strong>of</strong>acSurg 43:789–7943. Ameerally P, McGurk M, Shaheen O (1996) Atypical ameloblastoma:report <strong>of</strong> 3 cases <strong>and</strong> a review <strong>of</strong> <strong>the</strong> literature. Br JOral Maxill<strong>of</strong>ac Surg 34:235–2394. Auclair PL, Cuenin P, Kratochvil FJ, Slater LJ, Ellis GL (1988)A clinical <strong>and</strong> histomorphologic comparison <strong>of</strong> <strong>the</strong> centralgiant cell granuloma <strong>and</strong> <strong>the</strong> giant cell tumor. Oral Surg OralMed Oral Pathol 66:197–2085. August M, Faquin W, Troulis M, Kaban L (2003) Clear cellodontogenic carcinoma. Evaluation <strong>of</strong> reported cases. J OralMaxill<strong>of</strong>ac Surg 61:580–6856. Baden E, Moskow BS, Moskow R (1968) Odontogenic gingivalepi<strong>the</strong>lial hamartoma. J Oral Surg 26:702–7147. Badger JV, Gardner DG (1997) The relationship <strong>of</strong> adamantinomatouscraniopharyngioma to ghost cell ameloblastoma<strong>of</strong> <strong>the</strong> jaws: a histopathologic <strong>and</strong> immunohistochemicalstudy. J Oral Pathol Med 26:349–3558. Barker BF (1999) Odontogenic myxoma. Semin Diagn Pathol16:297–3019. Barrett AW, Morgan M, Ramsay AD, Farthing PM, NewmanL, Speight PM (2002) A clinicopathologic <strong>and</strong> immunohistochemicalanalysis <strong>of</strong> melanotic neuroectodermal tumor <strong>of</strong>infancy. Oral Surg Oral Med Oral Pathol Oral Radiol Endod93:688–69810. Barron RP, Kainulainen VT, Forrest CR, Krafchik B, MockD, S<strong>and</strong>or GKB (2002) Tuberous sclerosis: clinicopathologicfeatures <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. J Craniomaxill<strong>of</strong>ac Surg30:361–36611. Brannon RB (1976) The odontogenic keratocyst. A clinicopathologicstudy <strong>of</strong> 312 cases. I. Clinical features. Oral SurgOral Med Oral Pathol 42:54–7212. Brannon RB (1977) The odontogenic keratocyst. A clinicopathologicstudy <strong>of</strong> 312 cases. II. Histopathologic features.Oral Surg Oral Med Oral Pathol 43:233–25513. Brannon RB, Fowler CB (2001) Benign fibro-osseous lesions:a review <strong>of</strong> current concepts. Adv Anat Pathol 8:126–14314. Brannon RB, Fowler CB, Carpenter WM, Corio RL (2002)Cementoblastoma: an innocuous neoplasm? A clinicopathologicstudy <strong>of</strong> 44 cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature with spe-


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 127cial emphasis on recurrence. Oral Surg Oral Med Oral PatholOral Radiol Endod 93:311–32015. Brannon RB, Goode K, Eversole LR, Carr RF (2002) The centralgranular cell odontogenic tumor: report <strong>of</strong> 5 new cases.Oral Surg Oral Med Oral Pathol Oral Radiol Endod 94:614–62116. Braunshtein E, Vered M, Taicher S, Buchner A (2003) Clearcell odontogenic carcinoma <strong>and</strong> clear cell ameloblastoma.A single clinicopathologic entity? A new case <strong>and</strong> comparativeanalysis <strong>of</strong> <strong>the</strong> literature. J Oral Maxill<strong>of</strong>ac Surg 61:1004–101017. Brinck U, Gunawan B, Schulten HJ, Pinzon W, Fischer U,Fuezesi L (2001) Clear cell odontogenic carcinoma with pulmonarymetastases resembling pulmonary meningo<strong>the</strong>liallikenodules. Virchows Arch 438:412–41718. Browne RM (1971) The odontogenic keratocyst. Histologicalfeatures <strong>and</strong> <strong>the</strong>ir correlation with clinical behavior. Br DentJ 131:249–25919. Buchner A (1991) The central (intraosseous) calcifying odontogeniccyst: an analysis <strong>of</strong> 215 cases. J Oral Maxill<strong>of</strong>ac Surg49:330–33920. Buchner A, Merrell PW, Hansen LS, Leider AS (1991) Peripheral(extraosseous) calcifying odontogenic cyst. Oral SurgOral Med Oral Pathol 72:265–27021. Buchner A, Hansen LS (1979) The histomorphologic spectrum<strong>of</strong> <strong>the</strong> gingival cyst in <strong>the</strong> adult. Oral Surg Oral MedOral Pathol 48:523–53922. Budnick SD (1976) Compound <strong>and</strong> complex odontomas.Oral Surg Oral Med Oral Pathol 42:501–50623. Calvo N, Alonso D, Prieto M, Junquera L (2002) Centralodontogenic fibroma granular cell variant: a case report <strong>and</strong>review <strong>of</strong> <strong>the</strong> literature. J Oral Maxill<strong>of</strong>ac Surg 60:1192–119424. Cataldo E, Berkman MD (1968) Cysts <strong>of</strong> <strong>the</strong> oral mucosa innewborns. Am J Dis Child 16:44–4825. Chambers PW, Schwinn CP (1979) Chordoma. A clinicopathologicstudy <strong>of</strong> metastasis. Am J Clin Pathol 72:765–77626. Chehade A, Daley TD, Wysocki GP, Miller AS (1994) Peripheralodontogenic keratocyst. Oral Surg Oral Med Oral Pathol77:494–49727. Cheng YSL, Wright JM, Walstad WR, Finn MD (2002) Calcifyingepi<strong>the</strong>lial odontogenic tumor showing microscopicfeatures <strong>of</strong> potential malignant behavior. Oral Surg OralMed Oral Pathol Oral Radiol Endod 93:287–29528. C<strong>of</strong>fin CM, Swanson PE, Wick MR, Dehner LP (1993) An immunohistochemicalcomparison <strong>of</strong> chordoma with renal cellcarcinoma, colorectal adenocarcinoma, <strong>and</strong> myxopapillaryependymoma: a potential diagnostic dilemma in <strong>the</strong> diminutivebiopsy. Mod Pathol 6:531–53829. Colli B, Al-Mefty O (2001) Chordomas <strong>of</strong> <strong>the</strong> craniocervicaljunction: follow-up review <strong>and</strong> prognostic factors. J Neurosurg95:933–94330. Corio RL, Goldblatt LI, Edwards, PA, Hartman KS (1987)Ameloblastic carcinoma: a clinicopathologic study <strong>and</strong> assessment<strong>of</strong> eight cases. Oral Surg Oral Med Oral Pathol64:570–57631. Dal-Cin PD, Sciot R, Fossion E, Damme B van, Berghe H v<strong>and</strong>en (1993) Chromosome abnormalities in cementifying fibroma.Cancer Genet Cytogenet 71:170–17232. Daley TD, Wysocki GP (1994) Peripheral odontogenic fibroma.Oral Surg Oral Med Oral Pathol 78:329–33633. Daley TD, Wysocki GP (1995) The small dentigerous cyst.Oral Surg Oral Med Oral Pathol Oral Radiol Endod 79:77–8134. Daley TD, Wysocki GP, Pringle GA (1994) Relative incidence<strong>of</strong> odontogenic tumors <strong>and</strong> oral <strong>and</strong> jaw cysts in a Canadianpopulation. Oral Surg Oral Med Oral Pathol 77:276–28035. Deron PB, Nikolovski N, Holl<strong>and</strong>er JC den, Spoelstra HA,Knegt PP (1996) Myxoma <strong>of</strong> <strong>the</strong> maxilla; a case with extremelyaggressive biologic behavior. <strong>Head</strong> <strong>Neck</strong> 18:459–46436. Dhir K, Sciubba J, Tufano RP (2003) Case report. Ameloblasticcarcinoma <strong>of</strong> <strong>the</strong> maxilla. Oral Oncol 39:736–74137. Dunlap CL (1999) Odontogenic fibroma. Semin Diagn Pathol16:293–29638. Dunlap C, Neville B, Vickers RA, O’Neil D, Barker B (1989)The Noonan syndrome/cherubism association. Oral SurgOral Med Oral Pathol 67:698–70539. Ebata K, Takeshi U, Tohnai I, Kaneda T (1992) Chondrosarcoma<strong>and</strong> osteosarcoma arising in polyostotic fibrous dysplasia.J Oral Maxill<strong>of</strong>ac Surg 50:761–76440. El-M<strong>of</strong>ty SK (2002) Psammomatoid <strong>and</strong> trabecular juvenileossifying fibroma <strong>of</strong> <strong>the</strong> crani<strong>of</strong>acial skeleton: two distinctclinicopathologic entities. Oral Surg Oral Med Oral PatholOral Radiol Endod 93:296–30441. Elzay RP (1982) Primary intraosseous carcinoma <strong>of</strong> <strong>the</strong> jaws.Review <strong>and</strong> update <strong>of</strong> odontogenic carcinomas. Oral SurgOral Med Oral Pathol 54:299–30342. Eversole LR (1999) Malignant epi<strong>the</strong>lial odontogenic tumors.Semin Diagn Pathol 16:317–32443. Eversole LR, Duffey DC, Powell NB (1995) Clear cell odontogeniccarcinoma. A clinicopathologic analysis. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 121:685–68944. Eversole LR, Sabes WR, Rovin S (1972) Fibrous dysplasia: anosologic problem in <strong>the</strong> diagnosis <strong>of</strong> fibro-osseous lesions <strong>of</strong><strong>the</strong> jaws. J Oral Pathol 1:180–22045. Fowler CB, Brannon RB (1989) The paradental cyst: a clinicopathologicstudy <strong>of</strong> six new cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.J Oral Maxill<strong>of</strong>ac Surg 47:243–24846. Friedrich RE, Siegert J, Donath K, Jäkel KT (2001) Recurrentameloblastic fibro-odontoma in a 10-year-old boy. J OralMaxill<strong>of</strong>ac Surg 59:1362–136647. Gardner DG (1996) Some current concepts on <strong>the</strong> pathology<strong>of</strong> ameloblastomas. Oral Surg Oral Med Oral Pathol Oral RadiolEndod 82:660–66948. Gardner DG, Morency R (1993) The gl<strong>and</strong>ular odontogeniccyst; a rare lesion that tends to recur. J Can Dent Assoc59:929–93049. Gollin SM, Storto PD, Malone PS, Barnes L, Washington JA,Chidambaram A, Janecka IP (1992) Cytogenetic abnormalitiesin an ossifying fibroma from a patient with bilateral retinoblastoma.Genes Chromosomes Cancer 4:146–15250. Gorlin RJ, Goltz RW (1960) Multiple nevoid basal cell epi<strong>the</strong>lioma,jaw cysts <strong>and</strong> bifid rib. A syndrome. N Engl J Med262:908–91251. Gottschalk D, Fehn M, Patt S, Saeger W, Kirchner T, AignerT (2001) Matrix gene expression analysis <strong>and</strong> cellular phenotypingin chordoma reveals focal differentiation pattern <strong>of</strong>neoplastic cells mimicking nucleus pulposus development.Am J Pathol 158:1571–157852. Greer RO, Johnson M (1988) Botryoid odontogenic cyst:clinicopathologic analysis <strong>of</strong> ten cases with three recurrences.J Oral Maxill<strong>of</strong>ac Surg 46:574–57953. Groot RH, Merkesteyn JPR van, Bras J (1996) Diffuse sclerosingosteomyelitis <strong>and</strong> florid osseous dysplasia. Oral SurgOral Med Oral Pathol Oral Radiol Endod 81:333–34254. Gurol M. Burkes EJ, Jacoway J (1995) Botryoid odontogeniccyst: analysis <strong>of</strong> 33 cases. J Periodontol 66:1069–107355. H<strong>and</strong>lers JP, Abrams AM, Melrose RJ, Danfort R (1991) Centralodontogenic fibroma. Clinicopathologic features <strong>of</strong> 19cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. J Oral Maxill<strong>of</strong>ac Surg49:46–5456. Hansen LS, Eversole LR, Green TL, Powell NB (1985) Clearcell odontogenic tumor – a new histologic variant with aggressivepotential. <strong>Head</strong> <strong>Neck</strong> Surg 8:115–12357. Heffelfinger MJ, Dahlin DC, MacCarry CS, Beabout JW(1973) Chordomas <strong>and</strong> cartilaginous tumors at <strong>the</strong> skullbase. Cancer 32:410–42058. Hicks MJ, Flaitz CM, Wong ME, McDaniel RK, Cagle PT(1994) Clear cell variant <strong>of</strong> calcifying epi<strong>the</strong>lial odontogenictumor: case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. <strong>Head</strong> <strong>Neck</strong>16:272–27759. Higuchi Y, Nakamura N, Tashiro H (1988) Clinicopathologicstudy <strong>of</strong> cemento-osseous dysplasia producing cysts <strong>of</strong> <strong>the</strong>m<strong>and</strong>ible – report <strong>of</strong> four cases. Oral Surg Oral Med OralPathol 65:339–34260. Hirschberg A, Dayan D, Horowitz I (1987) Dentinogenicghost cell tumor. Int J Oral Maxill<strong>of</strong>ac Surg 16:620–625


128 P.J. Slootweg461. Hirschberg A, Kaplan I, Buchner A (1994) Calcifying odontogeniccyst associated with odontoma: a possible separateentity (odontocalcifying odontogenic cyst). J Oral Maxill<strong>of</strong>acSurg 52:555–55862. Horner K, Forman GH (1988) Atypical simple bone cysts <strong>of</strong><strong>the</strong> jaws. II. A possible association with benign fibro-osseous(cemental) lesions <strong>of</strong> <strong>the</strong> jaws. Clin Radiol 39:59–6363. Houston GD, Fowler CB (1997) Extraosseous calcifying epi<strong>the</strong>lialodontogenic tumor. Report <strong>of</strong> two cases <strong>and</strong> review <strong>of</strong><strong>the</strong> literature. Oral Surg Oral Med Oral Pathol Oral RadiolEndod 83:577–58364. Hruban RH, May M, Marcove RC, Huvos AG (1990) Lumbosacralchordoma with high-grade malignant cartilaginous<strong>and</strong> spindle cell components. Am J Surg Pathol 14:384–38965. Ide F, Saito I (2003) Letter to <strong>the</strong> editor. Many faces <strong>of</strong> odontogenickeratocyst. Oral Oncol 39:204–20566. Ide F, Sakshita H, Kusama K (2002) Ameloblastomatoid, centralodontogenic fibroma: an epi<strong>the</strong>lium-rich variant. J OralPathol Med 31:612–61467. Johnson LM, Sapp JP, McIntire DN (1994) Squamous cellcarcinoma arising in a dentigerous cyst. J Oral Maxill<strong>of</strong>acSurg 52:987–99068. Kapadia SB, Frisman DM, Hitchcock CL, Popek EJ (1993)Melanotic neuroectodermal tumor <strong>of</strong> infancy. Clinicopathological,immunohistochemical, <strong>and</strong> flow cytometric study.Am J Surg Pathol 17:566–57369. Kenney JN, Kaugars GE, Abbey LM (1989) Comparison between<strong>the</strong> peripheral ossifying fibroma <strong>and</strong> peripheral odontogenicfibroma. J Oral Maxill<strong>of</strong>ac Surg 47:378–38270. Khanna JN, Andrade NN (1993) Giant ossifying fibroma.Case report on a bimaxillary presentation. Int J Oral Maxill<strong>of</strong>acSurg 21:233–23571. Kim J, Ellis GL (1993) Dental follicular tissue: misinterpretationas odontogenic tumors. J Oral Maxill<strong>of</strong>ac Surg 51:762–76772. Kimonis VE, Goldstein AM, Pastakia B, Yang ML, Kase R,DiGiovanna JJ, Bale AE, Bale SJ (1997) Clinical manifestationsin 105 patients with nevoid basal cell syndrome. Am JMed Genet 69:299–30873. Kramer IRH, Pindborg JJ, Shear M (1992) Histological typing<strong>of</strong> odontogenic tumours, 2nd edn. Springer, Berlin HeidelbergNew York, pp 10–4274. Kratochvil FJ, Brannon RB (1993) Cartilage in <strong>the</strong> walls <strong>of</strong>odontogenic keratocysts. J Oral Pathol Med 22:282–28575. Kumar M, Fasanmade A, Barrett AW, Mack G, Newman L,Hyde NC (2003) Metastasising clear cell odontogenic carcinoma:a case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Oral Oncol39:190–19476. Kunze E, Donath K, Luhr HG, Engelhardt W, DeVivie R(1985) Biology <strong>of</strong> metastasizing ameloblastoma. Pathol ResPract 180:526–53577. Laughlin EH (1989) Metastasizing ameloblastoma. Cancer64:776–78078. Ledesma CM, Taylor AM, Romero de Leon E, Piedra GarzaM de la, Jaukin PG, Robertson JP (1993) Adenomatoid odontogenictumour with features <strong>of</strong> calcifying epi<strong>the</strong>lial odontogenictumour. (The so-called combined epi<strong>the</strong>lial odontogenictumour.) Clinico-pathological report <strong>of</strong> 12 cases. OralOncol Eur J Cancer 29B:221–22479. Leider AS, Jonker LA, Cook HE (1989) Multicentric familialsquamous odontogenic tumor. Oral Surg Oral Med OralPathol 68:175–18180. Levy BA (1973) Ghost cells <strong>and</strong> odontomas. Oral Surg OralMed Oral Pathol 36:851–85581. Li TJ, Yu SF (2003) Clinicopathologic spectrum <strong>of</strong> <strong>the</strong> socalledcalcifying odontogenic cysts. A study <strong>of</strong> 21 intraosseouscases with reconsideration <strong>of</strong> <strong>the</strong> terminology <strong>and</strong> classification.Am J Surg Pathol 27:372–38482. Lombardi T, Lock C, Samson J, Odell EW (1995) S100, α-smooth muscle actin <strong>and</strong> cytokeratin 19 immunohistochemistryin odontogenic <strong>and</strong> s<strong>of</strong>t tissue myxomas. J Clin Pathol48:759–76283. Lopez-Rios F, Lassaletta-Atienza L, Domingo-Carrasco C,Martinez-Tello FJ (1997) Nasolabial cyst: report <strong>of</strong> a casewith extensive apocrine change. Oral Surg Oral Med OralPathol Oral Radiol Endod 84:404–40684. Lu Y, Mock D, Takata T, Jordan RC (1999) Odontogenic ghostcell carcinoma: report <strong>of</strong> four new cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.J Oral Pathol Med 28:323–32985. Lukinmaa PL, Hietanen J, Anttinen J, Ahonen P (1990) Contiguousenlarged dental follicles with histologic features resembling<strong>the</strong> WHO type <strong>of</strong> odontogenic fibroma. Oral SurgOral Med Oral Pathol 70:313–31786. MacLeod RI, Fanibunda KB, Soames JV (1985) A pigmentedodontogenic keratocyst. Br J Oral Maxill<strong>of</strong>ac Surg 23:216–21987. MacLeod RI, Soames JV (1988) Squamous cell carcinoma inan odontogenic keratocyst. Br J Oral Maxill<strong>of</strong>ac Surg 26:52–5788. Magnusson B, Borrman H (1995) The paradental cyst: a clinicopathologicstudy <strong>of</strong> 26 cases. Swed Dent J 19:1–789. Maiorano E, Altini M, Viale G, Piatelli A, Favia G (2001)Clear cell odontogenic carcinoma. Report <strong>of</strong> two cases <strong>and</strong>review <strong>of</strong> <strong>the</strong> literature. Am J Clin Pathol 116:107–11490. Makowski GJ, McGuff S, Sickels JE van (2001) Squamouscell carcinoma in a maxillary odontogenic keratocyst. J OralMaxill<strong>of</strong>ac Surg 59:76–8091. Manojlovic S, Grguveric J, Knezevic G, Kruslin B (1997)Gl<strong>and</strong>ular odontogenic cyst: a case report <strong>and</strong> clinicopathologicanalysis <strong>of</strong> <strong>the</strong> relationship to central mucoepidermoidcarcinoma. <strong>Head</strong> <strong>Neck</strong> 19:227–23192. Mehta BS, Grewal GS (1963) Osteoma <strong>of</strong> <strong>the</strong> paranasal sinusesalong with a case <strong>of</strong> an orbito-ethmoid osteoma. J LaryngolOtol 77:601–61093. Miller R, Longo J, Houston G (1988) Surgical ciliated cyst <strong>of</strong><strong>the</strong> maxilla. J Oral Maxill<strong>of</strong>ac Surg 46:310–32294. Monteleone L, McLellan MS (1964) Epstein’s pearls (Bohn’snodules) <strong>of</strong> <strong>the</strong> palate. J Oral Surg 22:301–30495. Morgan PR, Johnson NW (1974) Histological, histochemical<strong>and</strong> ultrastructural studies on <strong>the</strong> nature <strong>of</strong> hyaline bodies inodontogenic cysts. J Oral Pathol 3:127–14796. Mosby EL, Russell D, Noren S, Barker BF (1998) Ameloblasticfibroma in a 7-week-old infant; a case report <strong>and</strong> review <strong>of</strong><strong>the</strong> literature. J Oral Maxill<strong>of</strong>ac Surg 56:368–37297. Mosqueda-Taylor A, Carlos-Bregni R, Ramirez-Amador V,Palma-Guzman JM, Esquivel-Bonilla D, Hern<strong>and</strong>ez-RojasLA (2002) Odontoameloblastoma. Clinico-pathologic study<strong>of</strong> three cases <strong>and</strong> critical review <strong>of</strong> <strong>the</strong> literature. Oral Oncol38:800–80598. Muller S, Parker DC, Kapadia SB, Budnick SD, Barnes L(1995) Ameloblastic fibrosarcoma <strong>of</strong> <strong>the</strong> jaws. A clinicopathologic<strong>and</strong> DNA analysis <strong>of</strong> five cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literaturewith discussion <strong>of</strong> its relationship to ameloblastic fibroma.Oral Surg Oral Med Oral Pathol Oral Radiol Endod79:469–47799. Müller H, Slootweg PJ (1985) The growth characteristics <strong>of</strong>multilocular ameloblastomas. A histological investigationwith some inferences with regard to operative procedures. JMaxill<strong>of</strong>ac Surg 13:224–230100. Müller H, Slootweg PJ (1986) Clear cell differentiation in anameloblastoma. J Maxill<strong>of</strong>ac Surg 14:158–160101. Müller H, Slootweg PJ (1988) A peculiar finding in Le Fort Iosteotomy. J Craniomaxill<strong>of</strong>ac Surg 16:238–239102. Muzio LL, Nocini PF, Favia G, Procaccini M, Mignogna MD(1996) Odontogenic myxoma <strong>of</strong> <strong>the</strong> jaws. A clinical, radiologic,immunohistochemical, <strong>and</strong> ultrastructural study. OralSurg Oral Med Oral Pathol Oral Radiol Endod 82:426–433103. Nastri AL, Wiesenfeld D, Radden BG, Eveson J, Scully C(1995) Maxillary ameloblastoma: a retrospective study <strong>of</strong> 13cases. Br J Oral Maxill<strong>of</strong>ac Surg 33:28–32104. Newman L, Howells GL, Coghlan KM, DiBase A, WilliamsDM (1995) Malignant ameloblastoma revisited. Br J OralMaxill<strong>of</strong>ac Surg 33:47–50105. Ng KH, Siar CH (2003) Odontogenic keratocyst with dentinoidformation. Oral Surg Oral Med Oral Pathol Oral RadiolEndod 95:601–606106. Nitta T, Endo T, Tsunoda A, Kadota Y, Matsumoto T, Sato K(1995) Melanotic neuroectodermal tumor <strong>of</strong> infancy: a molecularapproach to diagnosis. J Neurosurg 83:145–148


Maxill<strong>of</strong>acial Skeleton <strong>and</strong> Teeth Chapter 4 129107. Nxumalo TN, Shear M (1992) Gingival cysts in adults. J OralPathol Med 21:309–313108. Odell EW, Lombardi T, Barrett AW, Morgan PR, SpeightPM (1997) Hybrid central giant cell granuloma <strong>and</strong> centralodontogenic fibroma-like lesions <strong>of</strong> <strong>the</strong> jaws. Histopathology30:165–171109. Odukoya O (1995) Odontogenic tumors: analysis <strong>of</strong> 289 Nigeriancases. J Oral Pathol Med 24:454–457110. Okada Y, Mochizuki K, Sigimural M, Noda Y, Mori M (1987)Odontogenic tumor with combined characteristics <strong>of</strong> adenomatoidodontogenic <strong>and</strong> calcifying epi<strong>the</strong>lial odontogenictumors. Pathol Res Pract 182:647–657111. Paulus W, Stöckel C, Krauss J, Sörensen N, Roggendorf W(1997) Odontogenic classification <strong>of</strong> craniopharyngiomas:a clinicopathological study <strong>of</strong> 54 cases. Histopathology30:172–176112. Petschler M, Stiller M, H<strong>of</strong>fmeister B,Witkowski R, Opitz C,Bill JS, Peters H (2003) Klinische und molekulargenetischeBefunde bei Familien mit Cherubismus über 3 Generationen.Mund Kiefer Gesichtschir 7:83–87113. Pettinato G, Manivel C, d’Amore ES, Jaszcz W, Gorlin RJ(1991) Melanotic neuroectodermal tumor <strong>of</strong> infancy. A reexamination<strong>of</strong> a histogenesis problem based on immunohistochemical,flow cytometric, <strong>and</strong> ultrastructural study <strong>of</strong> 10cases. Am J Surg Pathol 15:233–245114. Philipsen HP (1956) Om keratocystes (kolesteatomer). I Kaeberne,T<strong>and</strong>laegebladet 60:963–980115. Philipsen HP, Reichart PA (1998) Unicystic ameloblastoma.A review <strong>of</strong> 193 cases from <strong>the</strong> literature. Oral Oncol 34:317–325116. Philipsen HP, Reichart PA (2000) Calcifying epi<strong>the</strong>lial odontogenictumour: biological pr<strong>of</strong>ile based on 181 cases from<strong>the</strong> literature. Oral Oncol 36:17–26117. Philipsen HP, Reichart PA, Nikai H, Takata T, Kudo Y (2001)Peripheral ameloblastoma: biological pr<strong>of</strong>ile based on 160cases from <strong>the</strong> literature. Oral Oncol 37:17–27118. Philipsen HP, Reichart PA, Praetorius F (1997) Mixed odontogenictumours <strong>and</strong> odontomas. Considerations on interrelationship.Review <strong>of</strong> <strong>the</strong> literature <strong>and</strong> presentation <strong>of</strong> 134cases <strong>of</strong> odontomas. Oral Oncol 33:86–99119. Philipsen HP, Reichart PA, Takata T (2001) Desmoplasticameloblastoma (including “hybrid” lesion <strong>of</strong> ameloblastoma).Biological pr<strong>of</strong>ile based on 100 cases from <strong>the</strong> literature<strong>and</strong> own files. Oral Oncol 37:455–460120. Philipsen HP, Reichart PA, Zhang KH, Nikai AH, Yu QX(1991) Adenomatoid odontogenic tumor: biologic pr<strong>of</strong>ilebased on 499 cases. J Oral Pathol Med 20:149–158121. Piatelli A, Rubini C, Goteri G, Fioroni M, Maiorano E (2003)Case report. Central granular cell odontogenic tumour. Report<strong>of</strong> <strong>the</strong> first malignant case <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.Oral Oncol 39:78–82122. Piecuch JF, Eisenberg E, Segal D, Carlson R (1980) Respiratoryepi<strong>the</strong>lium as an integral part <strong>of</strong> an odontogenic keratocyst:report <strong>of</strong> a case. J Oral Surg 38:445–447123. Poll<strong>and</strong>t K, Engels C, Kaiser E, Werner M, Delling G (2001)Gsα gene mutations in monostotic fibrous dysplasia <strong>of</strong> bone<strong>and</strong> fibrous dysplasia-like low-grade central osteosarcoma.Virchows Arch 439:170–175124. Praetorius F, Hjorting-Hansen E, Gorlin RJ, Vickers RA(1981) Calcifying odontogenic cyst. Range, variations <strong>and</strong>neoplastic potential. Acta Odontol Sc<strong>and</strong> 39:227–240125. Radner H, Katenkamp D, Reifenberger G, Deckert M, PietschT, Wiestler OD (2001) New developments in <strong>the</strong> pathology<strong>of</strong> skull base tumors. Virchows Arch 438:321–335126. Ramer M, Montazem A, Lane SL, Lumerman H (1997) Gl<strong>and</strong>ularodontogenic cyst. Report <strong>of</strong> a case <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.Oral Surg Oral Med Oral Pathol Oral Radiol Endod84:54–57127. Regezi JA, Kerr DA, Courtney RM (1978) Odontogenic tumors:analysis <strong>of</strong> 706 cases. J Oral Surg 36:771–778128. Reichart PA, Philipsen HP (2003) Entzündliche paradentaleZyste. Bericht von 6 Fällen. Mund Kiefer Gesichtschir 7:171–174129. Reichart PA, Philipsen HP (2003) Revision der WHO-Klassifikationodontogener Tumoren von 1992. Ein Vorschlag.Mund Kiefer Gesichtschir 7:88–93130. Reichart PA, Philipsen HP, Sonner S (1995) Ameloblastoma:biological pr<strong>of</strong>ile <strong>of</strong> 3677 cases. Oral Oncol Eur J Cancer31B:86–99131. Rosenberg AE, Brown GA, Bhan AK, Lee JM (1994) Chondroidchordoma – a variant <strong>of</strong> chordoma. A morphologic <strong>and</strong>immunohistochemical study. Am J Clin Pathol 101:36–41132. Rosenberg AE, Nielsen GP, Keel SB, Renard LG, Fitzek, MunzenderJE, Liebsch NJ (1999) Chondrosarcoma <strong>of</strong> <strong>the</strong> base <strong>of</strong><strong>the</strong> skull: a clinicopathologic study <strong>of</strong> 200 cases with emphasison its distinction from chordoma. Am J Surg Pathol23:1370–1378133. Rosenstein T, Pogrel MA, Smith RA, Regezi JA (2001) Cysticameloblastoma – behavior <strong>and</strong> treatment <strong>of</strong> 21 cases. J OralMaxill<strong>of</strong>ac Surg 59:1311–1316134. Ruggieri M, Pavone V, Polizzi A, Albanese S, Magro G, MerinoM, Duray PH (1999) Unusual form <strong>of</strong> recurrent giantcell granuloma <strong>of</strong> <strong>the</strong> m<strong>and</strong>ible <strong>and</strong> lower extremities in apatient with neur<strong>of</strong>ibromatosis type 1. Oral Surg Oral MedOral Pathol Oral Radiol Endod 87:67–72135. Saied-Al-Naief NAH, Lumerman H, Ramer M, Kopp W,Kringstein GJ, Persenchino F, Torno R (1997) keratoameloblastoma<strong>of</strong> <strong>the</strong> maxilla. A case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.Oral Surg Oral Med Oral Pathol Oral Radiol Endod84:535–539136. Saito Y, Hoshina Y, Nagamine T, Nakajima T, Suzuki M,Hayashi T (1992) Simple bone cyst. A clinical <strong>and</strong> histopathologicstudy <strong>of</strong> fifteen cases. Oral Surg Oral Med Oral Pathol74:487–491137. Samy LL, Mostafa H (1971) Osteomata <strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasalsinuses with a report <strong>of</strong> twenty-one cases. J LaryngolOtol 85:449–469138. Sawyer JR, Tryka AF, Bell JM, Boop FA (1995) Nonr<strong>and</strong>omchromosome breakpoints at Xq26 <strong>and</strong> 2q33 characterize cemento-ossifyingfibromas <strong>of</strong> <strong>the</strong> orbit. Cancer 76:1853–1859139. Schafer DR, Thompson LD, Smith BC, Wenig BM (1998) Primaryameloblastoma <strong>of</strong> <strong>the</strong> sinonasal tract: a clinicopathologicstudy <strong>of</strong> 24 cases. Cancer 82:667–674140. Schneider LC, Mesa LM (1990) Differences between floridosseous dysplasia <strong>and</strong> chronic diffuse sclerosing osteomyelitis.Oral Surg Oral Med Oral Pathol 70:308–312141. Schneider LC, Mesa ML, Fraenkel D (1988) Osteoporoticbone marrow defect: radiographic features <strong>and</strong> pathogenicfactors. Oral Surg Oral Med Oral Pathol 65:127–129142. Schwartz-Arad D, Lustmann J, Ulmansky M (1990) Squamousodontogenic tumor. Review <strong>of</strong> <strong>the</strong> literature <strong>and</strong> casereport. Int J Oral Maxill<strong>of</strong>ac Surg 19:327–330143. Scott J, Wood GD (1989) Aggressive calcifying odontogeniccyst – a possible variant <strong>of</strong> ameloblastoma. Br J Oral Maxill<strong>of</strong>acSurg 27:53–59144. Shah RV, Jambhekhar NA, Rana DN, Raje NS, AlbuqurqueKV, Mistry RC, Desai PB, Advani SH (1994) Melanotic neuroectodermaltumor <strong>of</strong> infancy: report <strong>of</strong> a case with ganglionicdifferentiation. J Surg Oncol 55:65–68145. Sharpe PT (2001) Neural crest <strong>and</strong> tooth morphogenesis.Adv Dent Res 15:4–7146. Shear M (1992) Cysts <strong>of</strong> <strong>the</strong> oral regions, 3rd edn. Wright,Oxford, p 6147. Shear M (2002) The aggressive nature <strong>of</strong> <strong>the</strong> odontogenickeratocyst: is it a benign cystic neoplasm? I. Clinical <strong>and</strong> earlyexperimental evidence <strong>of</strong> aggressive behaviour. Oral Oncol38:219–226148. Shear M (2002) The aggressive nature <strong>of</strong> <strong>the</strong> odontogenickeratocyst: is it a benign cystic neoplasm? II. Proliferation<strong>and</strong> genetic studies. Oral Oncol 38:323–331149. Shear M (2002) The aggressive nature <strong>of</strong> <strong>the</strong> odontogenickeratocyst: is it a benign cystic neoplasm? III. Immunocytochemistry<strong>of</strong> cytokeratin <strong>and</strong> o<strong>the</strong>r epi<strong>the</strong>lial markers. OralOncol 38:407–415150. Shear M, Pindborg JJ (1975) Microscopic features <strong>of</strong> <strong>the</strong> lateralperiodontal cyst. Sc<strong>and</strong> J Dent Res 83:103–110


130 P.J. Slootweg4151. Simko EJ, Brannon RB, Eibling DE (1998) Ameloblastic carcinoma<strong>of</strong> <strong>the</strong> m<strong>and</strong>ible. <strong>Head</strong> <strong>Neck</strong> 20:654–659152. Slater LJ (1999) Odontogenic sarcoma <strong>and</strong> carcinosarcoma.Semin Diagn Pathol 16:325–332153. Slootweg PJ (1981) An analysis <strong>of</strong> <strong>the</strong> interrelationship <strong>of</strong> <strong>the</strong>mixed odontogenic tumors – ameloblastic fibroma, ameloblasticfibro-odontoma, <strong>and</strong> <strong>the</strong> odontomas. Oral Surg OralMed Oral Pathol 51:266–276154. Slootweg PJ (1987) Carcinoma arising from reduced enamelepi<strong>the</strong>lium. J Oral Pathol 16:479–482155. Slootweg PJ (1991) Bone <strong>and</strong> cementum as stromal featuresin Pindborg tumor. J Oral Pathol Med 20:93–95156. Slootweg PJ (1992) Cementoblastoma <strong>and</strong> osteoblastoma: acomparison <strong>of</strong> histologic features. J Oral Pathol Med 21:385–389157. Slootweg PJ (1992) Heterologous tissue elements in melanoticneuroectodermal tumor <strong>of</strong> infancy. J Oral Pathol Med21:90–92158. Slootweg PJ (1996) Maxill<strong>of</strong>acial fibro-osseous lesions: classification<strong>and</strong> differential diagnosis. Semin Diagn Pathol13:104–112159. Slootweg PJ (2002) Malignant odontogenic tumors; an overview.Mund Kiefer Gesichtschir 6:295–302160. Slootweg PJ, Müller H (1983) Central fibroma <strong>of</strong> <strong>the</strong> jaw,odontogenic or desmoplastic. Oral Surg Oral Med OralPathol 56:61–70161. Slootweg PJ, Wittkampf ARM (1986) Myxoma <strong>of</strong> <strong>the</strong> jaws.An analysis <strong>of</strong> 15 cases. J Maxill<strong>of</strong>ac Surg 14:46–52162. Soames JV (1982) A pigmented calcifying odontogenic cyst.Oral Surg Oral Med Oral Pathol 53:395–400163. Stolovitzky JP, Waldron CA, McConnel FMS (1994) Giantcell lesions <strong>of</strong> <strong>the</strong> maxilla <strong>and</strong> paranasal sinuses. <strong>Head</strong> <strong>Neck</strong>16:143–148164. Su L, Wea<strong>the</strong>rs DR, Waldron CA (1997) Distinguishing features<strong>of</strong> focal cemento-osseous dysplasias <strong>and</strong> cemento-ossifyingfibromas. II. A clinical <strong>and</strong> radiologic spectrum <strong>of</strong> 316cases. Oral Surg Oral Med Oral Pathol Oral Radiol Endod84:540–549165. Suarez PA, Batsakis JG, El-Naggar AJ (1996) <strong>Pathology</strong> consultation.Don’t confuse dental s<strong>of</strong>t tissues with odontogenictumors. Ann Otol Rhinol Laryngol 105:490–494166. Suljak JP, Bohay RN, Wysocki GP (1998) Lateral periodontalcyst: a case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. J Can Dent Assoc64:58–51167. Summerlin DJ, Tomich CE (1994) Focal cemento-osseousdysplasia: a clinicopathologic study <strong>of</strong> 221 cases. Oral SurgOral Med Oral Pathol 78:611–620168. Swanson KS, Kaugars GE, Gunsolley JC (1991) Nasopalatineduct cyst: an analysis <strong>of</strong> 334 cases. J Oral Maxill<strong>of</strong>ac Surg49:268–271169. Takahashi H, Fujita S, Okabe H (1991) Immunohistochemicalinvestigation in odontogenic myxoma. J Oral Pathol Med20:114–119170. Takeda Y (1999) Review. Ameloblastic fibroma <strong>and</strong> relatedlesions: current pathologic concept. Oral Oncol 35:535–540171. Thesleff I, Keränen S, Jernvall J (2001) Enamel knots as signalingcenters linking tooth morphogenesis <strong>and</strong> odontoblastdifferentiation. Adv Dent Res 15:14–18172. Thompson LD, Gyure KA (2000) Extracranial sinonasal tractmeningiomas: a clinicopathologic study <strong>of</strong> 30 cases with review<strong>of</strong> <strong>the</strong> literature. Am J Surg Pathol 24:640–650173. Toida M (1998) So-called calcifying odontogenic cyst: review<strong>and</strong> discussion on <strong>the</strong> terminology <strong>and</strong> classification. J OralPathol Med 27:49–52174. Tomich CE (1999) Benign mixed odontogenic tumors. SeminDiagn Pathol 16:308–316175. Venness MJ, Morgan G, Collins AP (2001) Calcifying epi<strong>the</strong>lialodontogenic (Pindborg) tumor with malignant transformation<strong>and</strong> metastatic spread. <strong>Head</strong> <strong>Neck</strong> 23:692–696176. Villiers Slabbert H de, Altini M (1991) Peripheral odontogenicfibroma: a clinicopathologic study. Oral Surg Oral MedOral Pathol 72:86–90177. Waldron CA, Koh ML (1990) Central mucoepidermoid carcinoma<strong>of</strong> <strong>the</strong> jaws: report <strong>of</strong> four cases with analysis <strong>of</strong> <strong>the</strong>literature <strong>and</strong> discussion <strong>of</strong> <strong>the</strong> relationship to mucoepidermoid,sialodontogenic <strong>and</strong> gl<strong>and</strong>ular odontogenic cysts. JOral Maxill<strong>of</strong>ac Surg 48:871–877178. Warter A, George-Diolombi G, Chazal M, Ango A (1990)Melanin in a dentigerous cyst <strong>and</strong> associated adenomatoidodontogenic tumor. Cancer 66:786–788179. Wenig BM, Vinh TN, Smirniotopoulos JG, Fowler CB, HoustonGD, Heffner DK (1995) Aggressive psammomatoid ossifyingfibromas <strong>of</strong> <strong>the</strong> sinonasal region: a clinicopathologicstudy <strong>of</strong> a distinct group <strong>of</strong> fibro-osseous lesions. Cancer76:1155–1165180. Wesley RK, Scannel T, Nathan LE (1984) Nasolabial cyst:presentation <strong>of</strong> a case with a review <strong>of</strong> <strong>the</strong> literature. J OralMaxill<strong>of</strong>ac Surg 42:188–192181. World Health Organization Classification <strong>of</strong> Tumours (2005)<strong>Pathology</strong> <strong>and</strong> genetics <strong>of</strong> tumours <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck.IARC, Lyon182. Williams TP, Connor FA Jr (1994) Surgical management<strong>of</strong> <strong>the</strong> odontogenic keratocyst: aggressive approach. J OralMaxill<strong>of</strong>ac Surg 52:964–966183. Williams SC, Peller PJ (1994) Gardner’s syndrome. Case report<strong>and</strong> discussion <strong>of</strong> <strong>the</strong> manifestations <strong>of</strong> <strong>the</strong> disorder.Clin Nucl Med 19:668–670184. Wilson D, Walker M, Aurora N, Moore S (2001) Ameloblastomawith mucous cell differentiation. Oral Surg Oral MedOral Pathol Oral Radiol Endod 91:576–578185. Woolgar JA, Rippin JW, Browne RM (1987) A comparativehistological study <strong>of</strong> odontogenic keratocysts in basal cellnaevus syndrome <strong>and</strong> non-syndrome patients. J Oral Pathol16:75–80186. Woolgar JA, Rippin JW, Browne RM (1987) The odontogenickeratocyst <strong>and</strong> its occurrence in <strong>the</strong> nevoid basal cell carcinomasyndrome. Oral Surg Oral Med Oral Pathol 64:727–730187. Wright JM (1979) Squamous odontogenic tumorlike proliferationsin odontogenic cysts. Oral Surg Oral Med Oral Pathol47:354–358188. Wright JM (1981) The odontogenic keratocyst: orthokeratinizedvariant. Oral Surg Oral Med Oral Pathol 51:609–618189. Young SK, Markowitz NR, Sullivan S, Seale TW, Hirschi R(1989) Familial gigantiform cementoma: classification <strong>and</strong>presentation <strong>of</strong> a large pedigree. Oral Surg Oral Med OralPathol 68:740–747190. Zwetyenga N, Pinsolle J, Rivel J, Majoufre-Lefebvre C, FaucherA, Pinsolle V (2001) Primary intraosseous carcinoma <strong>of</strong><strong>the</strong> jaws. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 127:794–797


Chapter 5Major <strong>and</strong> MinorSalivary Gl<strong>and</strong>sS. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5Contents5.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 1325.1.1 Normal Salivary Gl<strong>and</strong>s . . . . . . . . . . . . . . . . . 1325.1.2 Developmental Disorders . . . . . . . . . . . . . . . . 1325.2 Obstructive Disorders . . . . . . . . . . . . . . . . . . 1325.2.1 Mucus Escape Reaction . . . . . . . . . . . . . . . . . . 1325.2.2 Chronic Sclerosing Sialadenitis<strong>of</strong> <strong>the</strong> Subm<strong>and</strong>ibular Gl<strong>and</strong> (Küttner Tumour) . . . 1335.3. Infections . . . . . . . . . . . . . . . . . . . . . . . . . . 1335.3.1 Bacteria, Fungi . . . . . . . . . . . . . . . . . . . . . . . 1335.3.2 Viruses . . . . . . . . . . . . . . . . . . . . . . . . . . . 1335.4 Miscellaneous Inflammatory Disorders . . . . . . . . 1335.5 Miscellaneous Non-Inflammatory Disorders . . . . . 1335.5.1 Necrotising Sialometaplasia(Salivary Gl<strong>and</strong> Infarction) . . . . . . . . . . . . . . . 1335.5.2 Sialadenosis . . . . . . . . . . . . . . . . . . . . . . . . 1335.5.3 Adenomatoid Hyperplasia<strong>of</strong> Mucous Salivary Gl<strong>and</strong>s . . . . . . . . . . . . . . . . 1345.5.4 Irradiation Changes . . . . . . . . . . . . . . . . . . . . 1345.5.5 Tissue ChangesFollowing Fine Needle Aspiration . . . . . . . . . . . 1345.6 Oncocytic Lesions . . . . . . . . . . . . . . . . . . . . . 1345.6.1 Focal <strong>and</strong> Diffuse Oncocytosis . . . . . . . . . . . . . 1345.6.2 Ductal Oncocytosis . . . . . . . . . . . . . . . . . . . . 1345.6.3 Multifocal Nodular Oncocytic Hyperplasia . . . . . . 1355.7 Cysts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1355.7.1 Salivary Polycystic Dysgenetic Disease . . . . . . . . . 1355.7.2 Mucoceles . . . . . . . . . . . . . . . . . . . . . . . . . 1355.7.3 Simple Salivary Duct Cysts . . . . . . . . . . . . . . . 1355.7.4 Lymphoepi<strong>the</strong>lial Cystic Lesions . . . . . . . . . . . . 1355.7.4.1 Benign Lymphoepi<strong>the</strong>lial Cyst . . . . . . . . . . . . . 1355.7.4.2 Cystic Lymphoid Hyperplasia <strong>of</strong> AIDS . . . . . . . . . 1365.7.5 Sclerosing Polycystic Sialadenopathy(Sclerosing Polycystic Adenosis) . . . . . . . . . . . . 1365.7.6 O<strong>the</strong>r Cysts . . . . . . . . . . . . . . . . . . . . . . . . . 1375.8 Benign Tumours . . . . . . . . . . . . . . . . . . . . . . 1375.8.1 Pleomorphic Adenoma . . . . . . . . . . . . . . . . . . 1375.8.1.1 Salivary Gl<strong>and</strong> Anlage Tumour(“Congenital Pleomorphic Adenoma”) . . . . . . . . . 1405.8.2 Benign Myoepi<strong>the</strong>lioma . . . . . . . . . . . . . . . . . 1405.8.3 Basal Cell Adenoma . . . . . . . . . . . . . . . . . . . . 1415.8.4 Warthin’s Tumour . . . . . . . . . . . . . . . . . . . . . 1425.8.5 Oncocytoma . . . . . . . . . . . . . . . . . . . . . . . . 1435.8.6 Canalicular Adenoma . . . . . . . . . . . . . . . . . . 1435.8.7 Sebaceous Adenoma . . . . . . . . . . . . . . . . . . . 1445.8.8 Sebaceous Lymphadenoma . . . . . . . . . . . . . . . . 1445.8.9 Ductal Papilloma . . . . . . . . . . . . . . . . . . . . . 1445.8.10 Cystadenoma . . . . . . . . . . . . . . . . . . . . . . . . 1445.9 Malignant Epi<strong>the</strong>lial Tumours . . . . . . . . . . . . . 1445.9.1 Acinic Cell Carcinoma . . . . . . . . . . . . . . . . . . 1445.9.2 Mucoepidermoid Carcinoma . . . . . . . . . . . . . . 1465.9.3 Adenoid Cystic Carcinoma . . . . . . . . . . . . . . . 1475.9.4 Polymorphous Low-Grade Adenocarcinoma . . . . . 1485.9.4.1 Cribriform Adenocarcinoma <strong>of</strong> <strong>the</strong> Tongue . . . . . . 1495.9.5 Epi<strong>the</strong>lial-Myoepi<strong>the</strong>lial Carcinoma . . . . . . . . . . 1505.9.6 Hyalinising Clear Cell Carcinoma . . . . . . . . . . . 1515.9.7 Basal Cell Adenocarcinoma . . . . . . . . . . . . . . . 1515.9.8 Myoepi<strong>the</strong>lial Carcinoma(Malignant Myoepi<strong>the</strong>lioma) . . . . . . . . . . . . . . 1525.9.9 Salivary Duct Carcinoma . . . . . . . . . . . . . . . . 1545.9.10 Oncocytic Carcinoma . . . . . . . . . . . . . . . . . . 1555.9.11 Malignancy in Pleomorphic AdenomaMalignant Mixed Tumour . . . . . . . . . . . . . . . . 1565.9.11.1 Carcinoma (True Malignant Mixed Tumour)Ex Pleomorphic Adenoma . . . . . . . . . . . . . . . . 1565.9.11.2 Carcinosarcoma Ex Pleomorphic Adenoma . . . . . . 1575.9.11.3 Metastasising Pleomorphic Adenoma . . . . . . . . . 1575.9.12 Sebaceous Carcinoma . . . . . . . . . . . . . . . . . . . 1585.9.13 Lymphoepi<strong>the</strong>lial Carcinoma . . . . . . . . . . . . . . 1585.9.14 Small Cell Carcinoma . . . . . . . . . . . . . . . . . . . 1585.9.15 Higher Grade Change in Carcinomas . . . . . . . . . 1595.9.16 Metastatic Malignancies . . . . . . . . . . . . . . . . . 1595.10 Hybrid Carcinoma . . . . . . . . . . . . . . . . . . . . 1605.11 Endodermal Sinus Tumour . . . . . . . . . . . . . . . 1605.12 Sialoblastoma . . . . . . . . . . . . . . . . . . . . . . . 1605.13 Alterations in Gene Expression<strong>and</strong> Molecular Derangementsin Salivary Gl<strong>and</strong> Carcinoma . . . . . . . . . . . . . . 1605.13.1 Predominantly Myoepi<strong>the</strong>lial Malignancies . . . . . . 1615.13.2 Predominantly Epi<strong>the</strong>lial Malignancies . . . . . . . . 1615.14 Benign <strong>and</strong> Malignant Lymphoid Infiltrates . . . . . 1625.14.1 Non-Autoimmune Lymphoid Infiltrates . . . . . . . . 1625.14.2 Benign Autoimmune Lymphoid Infiltrates . . . . . . 1625.14.3 Malignant Lymphoma . . . . . . . . . . . . . . . . . . 1635.15 O<strong>the</strong>r Tumours . . . . . . . . . . . . . . . . . . . . . . 1635.16 Unclassified Tumours . . . . . . . . . . . . . . . . . . . 163References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164


132 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5.1 Introduction5.1.1 Normal Salivary Gl<strong>and</strong>s5The salivary gl<strong>and</strong>s include paired major gl<strong>and</strong>s (parotid,subm<strong>and</strong>ibular <strong>and</strong> sublingual) <strong>and</strong> minor gl<strong>and</strong>sthroughout <strong>the</strong> upper aerodigestive tract.The cellular component comprises serous <strong>and</strong> mucousacinar <strong>and</strong> ductal epi<strong>the</strong>lial cells, myoepi<strong>the</strong>lialcells <strong>and</strong> connective tissue components (e.g. fat, fibroustissue, nerves <strong>and</strong> blood vessels). The parotidgl<strong>and</strong>s consist <strong>of</strong> predominantly serous acini, <strong>the</strong> subm<strong>and</strong>ibulargl<strong>and</strong>s <strong>of</strong> mixed, serous <strong>and</strong> mucous acini,while <strong>the</strong> sublingual gl<strong>and</strong>s contain mainly mucousacini. Minor salivary gl<strong>and</strong>s also have mixed serous<strong>and</strong> mucous acini in varying proportions.Of particular interest are <strong>the</strong> myoepi<strong>the</strong>lial cells.They are a normal constituent <strong>of</strong> <strong>the</strong> major <strong>and</strong> minorsalivary gl<strong>and</strong>s, <strong>and</strong> are believed to have contractileproperties that assist in <strong>the</strong> secretion <strong>of</strong> saliva. Similarcells are also found in <strong>the</strong> breast, tracheo-bronchial<strong>and</strong> sweat gl<strong>and</strong>s. They are plentiful in <strong>the</strong> salivaryacini <strong>and</strong> intercalated ducts, but much less so in <strong>the</strong>larger excretory ducts <strong>of</strong> <strong>the</strong> major gl<strong>and</strong>s. Microscopicexamination shows that myoepi<strong>the</strong>lial cells are thin<strong>and</strong> spindle-shaped <strong>and</strong> situated between <strong>the</strong> basementmembrane <strong>and</strong> epi<strong>the</strong>lial cells, <strong>and</strong> ultrastructurally<strong>the</strong>y are seen to possess a number <strong>of</strong> cytoplasmicprocesses that extend between <strong>and</strong> over <strong>the</strong> acinar<strong>and</strong> ductal lining cells. They display features <strong>of</strong> bothsmooth muscle <strong>and</strong> epi<strong>the</strong>lium, such as numerous micr<strong>of</strong>ilamentswith focal densities in <strong>the</strong> cytoplasmicprocesses, <strong>and</strong> desmosomes that attach <strong>the</strong> myoepi<strong>the</strong>lialto <strong>the</strong> epi<strong>the</strong>lial cells [62]. Similarly, immunohistochemistryshows that myoepi<strong>the</strong>lial cells stainstrongly with alpha smooth muscle actin (SMA), calponin,smooth muscle myosin heavy chain (SMMHC)[164], h-caldesmon [74], S-100 protein [114] as well aswith some cytokeratins (e.g. subtype 14). Maspin, p63[8, 166] <strong>and</strong> CD 10 [143, 183] have recently been describedas markers <strong>of</strong> breast myoepi<strong>the</strong>lial cells, <strong>and</strong>may have a role in identifying <strong>the</strong>ir salivary equivalents.Preliminary studies show that p63 may well havepractical value [166]. Scattered nests <strong>of</strong> sebaceous cellscan be seen in normal parotid <strong>and</strong> minor salivarygl<strong>and</strong>s [62].Serial sectioning has shown an average <strong>of</strong> 20 lymphnodes within each parotid [67], <strong>and</strong> <strong>the</strong>y may be affectedby inflammatory processes <strong>and</strong> neoplasms, bothprimary <strong>and</strong> metastatic. Their presence may hamperhistologic evaluation <strong>of</strong> parotid gl<strong>and</strong> lesions [6].Fig. 5.1. Extravasation mucocele (mucous escape reaction): mucin-filledcavity lined with granulation tissue <strong>and</strong> macrophages5.1.2 Developmental DisordersAgenesis, aplasia, hypoplasia <strong>and</strong> atresia <strong>of</strong> <strong>the</strong> mainducts are all extremely rare. In contrast, intra-parotidnodal heterotopias are very common [129], <strong>and</strong> epi<strong>the</strong>lialtumours may arise from <strong>the</strong>m [175]. Extranodalheterotopia is rare, <strong>and</strong> can be subdivided into high(involvement <strong>of</strong> <strong>the</strong> ear, pituitary, m<strong>and</strong>ible, etc.) or lowforms (lower neck, thyroid).Accessory parotid gl<strong>and</strong>s comprising salivary tissueseparate from <strong>the</strong> main gl<strong>and</strong>, adjacent to Stenson’sduct, are found in 20% <strong>of</strong> people.5.2 Obstructive Disorders5.2.1 Mucus Escape ReactionThis forms an extravasation mucocele, which is definedas <strong>the</strong> pooling <strong>of</strong> mucus in <strong>the</strong> connective tissue in a cavitynot lined with epi<strong>the</strong>lium. Most patients are under30 years <strong>of</strong> age, <strong>and</strong> <strong>the</strong> minor gl<strong>and</strong>s are most <strong>of</strong>ten affected.The incidence by site is lower lip 65%, palate 4%,buccal mucosa 10%, <strong>and</strong> (in <strong>the</strong> major gl<strong>and</strong>s) parotid0.6%, subm<strong>and</strong>ibular 1.2% <strong>and</strong> sublingual 1.1%. Thepathogenesis is traumatic severance <strong>of</strong> a duct, leadingto mucus pooling. It presents in <strong>the</strong> lip as a raised, <strong>of</strong>tenblue, dome shaped swelling <strong>of</strong> <strong>the</strong> mucosa, usually2–10 mm in diameter, but it is generally larger in <strong>the</strong>sublingual gl<strong>and</strong> in <strong>the</strong> floor <strong>of</strong> <strong>the</strong> mouth where it isknown as a ranula. Microscopy shows a well-definedmucin-filled cavity lacking an epi<strong>the</strong>lial lining, but linedwith granulation tissue <strong>and</strong> macrophages (Fig. 5.1).


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 1335.2.2 Chronic Sclerosing(Atrophic) Sialadenitis<strong>of</strong> <strong>the</strong> Subm<strong>and</strong>ibular Gl<strong>and</strong>( Küttner Tumour)In most if not all cases, this is secondary to calculi in <strong>the</strong>excretory ducts <strong>of</strong> <strong>the</strong> major salivary gl<strong>and</strong>s, particularly<strong>the</strong> subm<strong>and</strong>ibular gl<strong>and</strong>. It can occur at any age,though <strong>the</strong> mean is 44 years. Patients present with pain<strong>and</strong>/or swelling associated with eating. Histology showsacinar atrophy <strong>and</strong> a chronic inflammatory infiltrate <strong>of</strong>variable intensity, but it can be heavy with lymphoid germinalcentre formation. The end stage <strong>of</strong> destruction <strong>of</strong><strong>the</strong> lobular architecture <strong>and</strong> scarring has been describedas salivary gl<strong>and</strong> cirrhosis [172].5.3 InfectionsFig. 5.2. Necrotising sialometaplasia. Most <strong>of</strong> <strong>the</strong> ducts <strong>and</strong> aciniare replaced by mature non-keratinising squamous epi<strong>the</strong>lium.The lobular architecture <strong>of</strong> <strong>the</strong> gl<strong>and</strong> is preserved5.3.1 Bacteria, FungiTuberculosis may involve <strong>the</strong> gl<strong>and</strong> itself or intra-parotidlymph nodes, <strong>and</strong> may present as a salivary mass.O<strong>the</strong>r granulomatous infections such as cat-scratch,fungus, sarcoid, leprosy, syphilis, tularaemia, Brucellaor toxoplasmosis can also occur in <strong>the</strong> salivarygl<strong>and</strong>s.5.3.2 VirusesSeveral viral diseases lead to infiltration by chronic inflammatorycells, but are rarely biopsied. This is especiallytrue <strong>of</strong> mumps, <strong>and</strong> also in Cytomegalovirus infection,which may involve <strong>the</strong> salivary gl<strong>and</strong>s as part <strong>of</strong>a systemic infection in ei<strong>the</strong>r <strong>the</strong> newborn or immunocompromisedadults, particularly those with AIDS. Thediagnosis is made by finding <strong>the</strong> characteristic enlargedcells with intranuclear inclusions [233]. O<strong>the</strong>r viral infectionsinclude Epstein-Barr Virus (EBV), Coxsackievirus <strong>and</strong> influenza virus, as well as human immunodeficiencyvirus (HIV). Several lesions may be seen in<strong>the</strong> salivary gl<strong>and</strong>s in patients with AIDS, in particularcystic lymphoid hyperplasia (see Sect. 5.7.4.2).5.4 Miscellaneous InflammatoryDisordersThere are a variety <strong>of</strong> non-infectious inflammatoryconditions such as sarcoidosis [230], Rosai-Dorfm<strong>and</strong>isease [75], xanthogranulomatous sialadenitis, amyloidosis[98] <strong>and</strong> Kimura’s disease [155]. They will not bediscussed here.5.5 Miscellaneous Non-InflammatoryDisorders5.5.1 Necrotising Sialometaplasia(Salivary Gl<strong>and</strong> Infarction)Necrotising sialometaplasia (salivary gl<strong>and</strong> infarction),is a benign, self-healing lesion, affecting especially <strong>the</strong>minor gl<strong>and</strong>s <strong>of</strong> <strong>the</strong> palate. Some cases follow surgery(about 1–8 weeks postoperatively) or even relatively minortrauma, such as from an ill-fitting denture, but <strong>of</strong>tenno predisposing factor is known, although <strong>the</strong> underlyingprocess is generally considered to be ischaemic [172].Microscopy shows lobular coagulative necrosis <strong>of</strong> acini(particularly in <strong>the</strong> early stages), squamous metaplasia<strong>of</strong> ducts, a chronic inflammatory cell infiltrate <strong>and</strong> pseudoepi<strong>the</strong>liomatoushyperplasia <strong>of</strong> <strong>the</strong> overlying surface[24]. There is a superficial resemblance to ei<strong>the</strong>r mucoepidermoidor squamous cell carcinoma, but <strong>the</strong> overalllobular architecture <strong>of</strong> <strong>the</strong> involved gl<strong>and</strong> is preserved.A similar reaction can be seen in <strong>the</strong> major gl<strong>and</strong>s aftersurgery or radio<strong>the</strong>rapy (Fig. 5.2) [17].5.5.2 SialadenosisSialadenosis [172] is a non-inflammatory process <strong>of</strong><strong>the</strong> salivary gl<strong>and</strong>s due to metabolic <strong>and</strong> secretorydisorders <strong>of</strong> <strong>the</strong> gl<strong>and</strong> parenchyma accompanied byrecurrent painless bilateral swelling <strong>of</strong> <strong>the</strong> parotidgl<strong>and</strong>s. The peak ages are <strong>the</strong> fifth <strong>and</strong> sixth decades[172]. It has been related to endocrine disorders (diabetesmellitis, ovarian <strong>and</strong> thyroid insufficiencies) aswell as autonomic nervous system dysfunction; <strong>the</strong>


134 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivounderlying process appears to be a disorder <strong>of</strong> salivarygl<strong>and</strong> innervation. It is also seen in malnutrition,chronic alcoholism, bulimia, liver cirrhosis <strong>and</strong> hasbeen linked to some drugs, such as antihypertensiveagents [62].It is rarely biopsied, but histologically <strong>the</strong>re is enlargement<strong>of</strong> <strong>the</strong> serous acinar cells (two or three times<strong>the</strong> normal size) <strong>and</strong> slight compression <strong>of</strong> <strong>the</strong> duct systemby <strong>the</strong> swollen acini.55.5.3 Adenomatoid Hyperplasia<strong>of</strong> Mucous Salivary Gl<strong>and</strong>sThis nodular hyperplastic lesion is usually asymptomatic,<strong>of</strong>ten being noted on routine dental examination.Most cases occur on <strong>the</strong> palate, but sometimes o<strong>the</strong>r minorgl<strong>and</strong>s can be involved [25]. It can affect all ages,although most patients are between 30 <strong>and</strong> 60 yearsold. There is a slight male predominance. Examinationreveals nodular mucosal swellings up to 30 mm in diameter.The aetiology is unknown, but possible relevantfactors include local trauma due to dentures or tobaccosmoking. The main histological feature <strong>of</strong> adenomatoidhyperplasia is <strong>the</strong> presence <strong>of</strong> hypertrophic <strong>and</strong>hyperplastic mucous lobules <strong>of</strong> minor salivary gl<strong>and</strong>s.Inflammation, fibrosis <strong>and</strong> cytological abnormality arenot usually seen.5.5.4 Irradiation ChangesSalivary gl<strong>and</strong>s are very sensitive to radiation, <strong>and</strong> xerostomiais a common complication. Acute radiationinjury <strong>of</strong> salivary gl<strong>and</strong>s manifests with swelling, vacuolation<strong>and</strong> necrosis <strong>of</strong> acinar cells. Initial acute inflammatoryresponse is later followed by chronic sclerosingsialadenitis characterised by loss <strong>of</strong> acini, focal squamousmetaplasia, <strong>and</strong> fibrosis. When all <strong>the</strong> salivarygl<strong>and</strong>s are involved, <strong>the</strong> loss <strong>of</strong> saliva is progressive <strong>and</strong>irreversible.5.5.5 Tissue ChangesFollowing Fine Needle AspirationFig. 5.3. Focal oncocytosis <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>. Some ducts <strong>and</strong>acinar cells show cytoplasmic oncocytic featuresFine needle aspiration (FNA) is an important techniquein <strong>the</strong> investigation <strong>of</strong> salivary disease, particularly tumours,but <strong>the</strong> procedure itself can have adverse effects,causing difficulties in histological assessment <strong>and</strong> evensimulating malignancy. The effects are classified as tissueinjury with repair, infarction <strong>and</strong> reactive pseudomalignantchanges [28]. Some or all <strong>of</strong> <strong>the</strong>se can occurin any tumour [126] including pleomorphic adenoma,but are most frequent in Warthin’s tumour, where infarctionmay be total <strong>and</strong> squamous metaplasia florid[55]. Possible causes include trauma by <strong>the</strong> needle [55]<strong>and</strong> an increased sensitivity <strong>of</strong> oncocytic cells to hypoxia[28].5.6 Oncocytic LesionsOncocytic change is where cells develop intensely eosinophilicgranular cytoplasm due, to increased numbers <strong>of</strong>mitochondria [180].5.6.1 Focal<strong>and</strong> Diffuse OncocytosisFoci <strong>of</strong> oncocytic metaplasia, usually <strong>of</strong> ducts, but occasionallyalso acini, occur with increasing frequencywith advancing age (Fig. 5.3). In contrast, diffuse oncocytosis<strong>of</strong> <strong>the</strong> parotid is extremely rare. Microscopicexamination shows oncocytic metaplasia <strong>of</strong> ducts <strong>and</strong>acini involving virtually <strong>the</strong> whole gl<strong>and</strong>. As with mosto<strong>the</strong>r oncocytic lesions, diffuse oncocytosis comprisestwo types <strong>of</strong> cells, light <strong>and</strong> dark. The former are large<strong>and</strong> round or polygonal <strong>and</strong> have finely granular, pinkcytoplasm <strong>and</strong> a single vesicular nucleus. The darkcells are usually more sparse <strong>and</strong> have deeply eosinophilic,compressed cytoplasm <strong>and</strong> densely hyperchromaticnuclei.5.6.2 Ductal OncocytosisOncocytic metaplasia <strong>of</strong> ducts <strong>of</strong>ten with cystic dilation(also known as oncocytic papillary cystadenoma) occursmainly in <strong>the</strong> minor gl<strong>and</strong>s, particularly <strong>the</strong> larynx, <strong>and</strong>


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 135only occasionally in <strong>the</strong> parotid (see Sect. 5.8.10). Thelesions are <strong>of</strong>ten multifocal <strong>and</strong> usually small, but canreach 30 mm in diameter.5.6.3 Multifocal NodularOncocytic HyperplasiaThis rare condition consists <strong>of</strong> nodules <strong>of</strong> varying size,composed <strong>of</strong> oncocytic cells, <strong>of</strong>ten with relatively clearcytoplasm. The nodules appear to engulf normal acinigiving a false impression <strong>of</strong> invasion, but <strong>the</strong>re is nostromal or o<strong>the</strong>r response by <strong>the</strong> acini. Multifocal nodularoncocytic hyperplasia (MNOH) can be mistaken fora clear cell neoplasm with satellite deposits when onenodule is much larger than <strong>the</strong> o<strong>the</strong>rs. MNOH can alsobe bilateral, <strong>and</strong> it has been reported to co-exist with apleomorphic adenoma, which itself showed oncocyticchange [20, 158].lobular architecture, <strong>and</strong> some lobules are affectedmore severely than o<strong>the</strong>rs. The cysts vary in size up toa few millimetres, <strong>and</strong> <strong>the</strong>y are irregular in shape <strong>and</strong><strong>of</strong>ten interconnect. The lining epi<strong>the</strong>lium is flat, cuboidalto low columnar, sometimes with an apocrinelikeappearance. The lumen contains secretion withspherical microliths. Remnants <strong>of</strong> salivary acini areseen between <strong>the</strong> cysts, <strong>and</strong> thick fibrous interlobularsepta are <strong>of</strong>ten prominent.5.7.2 MucocelesA mucocele is defined as <strong>the</strong> pooling <strong>of</strong> mucus in a cysticcavity [62]. Two types are recognised – extravasation<strong>and</strong> retention; extravasation mucocele is described inSect. 5.2.1. Retention cysts can occur at any age, <strong>and</strong> <strong>the</strong>mucus pool is within an epi<strong>the</strong>lium-lined cavity, likelyto be a dilated excretory duct.5.7 CystsNon-neoplastic cysts <strong>and</strong> pseudocysts accounted forabout 6% <strong>of</strong> all lesions <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s in <strong>the</strong> Hamburgregistry <strong>of</strong> salivary neoplasms <strong>and</strong> tumour-like lesions[172]. They can be classified as:1. Dysgenetic, e.g. polycystic dysgenetic disease;2. Acquired cysts lined with epi<strong>the</strong>lium, e.g. lymphoepi<strong>the</strong>lialcystic lesions, duct cysts;3. Pseudocysts without an epi<strong>the</strong>lial lining, e.g. extravasationmucocele, including ranula;4. Cystic change in neoplasms, e.g. Warthin, variants<strong>of</strong> mucoepidermoid <strong>and</strong> acinic cell carcinomas, lymphoepi<strong>the</strong>lialsialadenitis (LESA), lymphoma, <strong>and</strong>rarely, pleomorphic adenoma;5. Miscellaneous o<strong>the</strong>r cysts.The commonest are mucoceles, including ranula (80%),parotid duct cysts (11%), lymphoepi<strong>the</strong>lial cystic lesions(7%) <strong>and</strong> dysgenetic cysts <strong>and</strong> congenital sialectasia (toge<strong>the</strong>r2%).5.7.1 Salivary PolycysticDysgenetic DiseaseThis very rare condition resembles cystic anomalies <strong>of</strong>o<strong>the</strong>r organs, such as <strong>the</strong> kidney, liver <strong>and</strong> pancreas,although no association has been described [62, 172,177]. Some cases are familial [207], <strong>and</strong> almost allcases occurred in females. Most patients present inchildhood, but some have not been recognised untiladulthood. It only affects <strong>the</strong> parotid gl<strong>and</strong>s, usuallybilaterally. Microscopically, <strong>the</strong> gl<strong>and</strong>s maintain <strong>the</strong>ir5.7.3 Simple Salivary Duct CystsSalivary duct cysts are acquired, <strong>and</strong> are due to dilatation<strong>of</strong> a salivary duct following obstruction, sometimesby a tumour [62]. They can occur at any age, althoughusually in patients over 30 years old. Most (85%) arisein <strong>the</strong> parotid <strong>and</strong> are unilateral <strong>and</strong> painless. They arewell-circumscribed, unilocular <strong>and</strong> up to 100 mm in diameter(usually 10 to 30 mm). They contain fluid that iswatery to viscous brown, occasionally with mucus. Thewall comprises dense fibrous tissue, 1–3 mm thick, <strong>and</strong><strong>the</strong>re is <strong>of</strong>ten mild to moderate chronic inflammation,although not <strong>the</strong> dense lymphoid infiltrate <strong>of</strong> a lymphoepi<strong>the</strong>lialcyst. The epi<strong>the</strong>lium is stratified squamous, ora single layer <strong>of</strong> cuboidal or columnar cells, with occasionalgoblet cells <strong>and</strong> oncocytes.5.7.4 Lymphoepi<strong>the</strong>lial Cystic LesionsSeven types <strong>of</strong> salivary lesions can be characterised bysingle or multiple epi<strong>the</strong>lial-lined cysts surrounded bylymphoid tissue including germinal centres: benignlymphoepi<strong>the</strong>lial cyst <strong>and</strong> cystic lymphoid hyperplasia<strong>of</strong> AIDS, in addition to Warthin’s tumour, LESA<strong>and</strong> mucosa-associated lymphoid tissue (MALT lymphoma)each with cystically dilated ducts, low-gradecyst-forming mucoepidermoid carcinoma with a heavylymphocytic response, <strong>and</strong> cystic metastases in intraparotidlymph nodes, each <strong>of</strong> which is discussed inSect. 5.14.


136 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5Fig. 5.4. Simple, benign lymphoepi<strong>the</strong>lial cyst. The cavity is linedwith columnar <strong>and</strong> cuboidal cells with scattered goblet cells. Thesurrounding tissue contains small lymphocytes <strong>and</strong> macrophages.Beyond this is a capsule <strong>and</strong> subcapsular space resembling that <strong>of</strong>a lymph nodeFig. 5.5. Sclerosing polycystic adenosis. Cystic ducts <strong>of</strong> varyingsize with stromal fibrosis, resembling fibrocystic disease <strong>and</strong> sclerosisadenosis <strong>of</strong> <strong>the</strong> breast. There is also proliferation <strong>of</strong> ducts <strong>and</strong>acini in a lobular pattern5.7.4.1 Benign Lymphoepi<strong>the</strong>lial CystBenign lymphoepi<strong>the</strong>lial cysts are thought to arise ei<strong>the</strong>rin intraparotid lymph nodes [60] or from remnants <strong>of</strong> <strong>the</strong>branchial apparatus [10]. There is no clinical associationwith Sjögren’s syndrome, <strong>and</strong> <strong>the</strong>y were described longbefore <strong>the</strong> AIDS epidemic. There is a slight male preponderance(1.6:1 in civilians in <strong>the</strong> Armed Forces Institute<strong>of</strong> <strong>Pathology</strong> [AFIP] series), <strong>and</strong> <strong>the</strong> mean age <strong>of</strong> onset is46 years (range 18 to 79) [113]. They are usually solitary,but can occasionally be bilateral. The average diameter is25 mm, but <strong>the</strong>y may reach 70 mm. Microscopy shows <strong>the</strong>lining epi<strong>the</strong>lium to be squamous, respiratory, cuboidal,columnar or a combination, <strong>and</strong> small numbers <strong>of</strong> gobletcells may also be present (Fig. 5.4). This lining is surroundedby abundant lymphoid tissue composed <strong>of</strong> smalllymphocytes, plasma cells <strong>and</strong> germinal centres; lymphoepi<strong>the</strong>liallesions are not a feature. Benign lymphoepi<strong>the</strong>lialcysts are not known to recur after surgical excision.5.7.4.2 Cystic LymphoidHyperplasia <strong>of</strong> AIDSA nodular or diffuse enlargement <strong>of</strong> particularly <strong>the</strong> parotidgl<strong>and</strong>s is <strong>of</strong>ten seen in HIV-positive patients – usuallybilaterally. Microscopic examination shows a denselymphoid infiltrate including follicular hyperplasia,sometimes displaying lysis <strong>of</strong> germinal centres <strong>and</strong> diminishedmantle zones. There is an elaborate dendriticreticulum cell network within which <strong>the</strong>re is evidence<strong>of</strong> active HIV replication, although <strong>the</strong> exact histogenesis<strong>of</strong> this lesion is not understood. Plasma cells (polytypic)are <strong>of</strong>ten numerous. The gl<strong>and</strong>ular parenchyma isatrophic, <strong>and</strong> multiple cystic spaces are seen, filled withmucoid or gelatinous fluid. The cysts are dilated ducts,<strong>and</strong> <strong>the</strong> lining sometimes shows squamous metaplasia.The cysts are infiltrated by lymphoid cells, includingvariable numbers <strong>of</strong> marginal zone B-cells, <strong>and</strong> in timelymphoepi<strong>the</strong>lial lesions are apparent.There is considerable morphological overlap withLESA [60, 91], but only a minority <strong>of</strong> patients exhibit<strong>the</strong> clinical features <strong>of</strong> Sjögren’s syndrome [105]. An importantpractical point is that this lesion can be <strong>the</strong> firstclinical manifestation <strong>of</strong> HIV disease, <strong>and</strong> thus histologicalidentification <strong>of</strong> it means a diagnosis <strong>of</strong> AIDS for<strong>the</strong> patient.The lymphoid infiltrate is polyclonal <strong>and</strong> generallydoes not progress to lymphoma, although patients withHIV disease are at risk <strong>of</strong> developing aggressive B-celllymphomas, most commonly <strong>of</strong> <strong>the</strong> Burkitt-type <strong>and</strong>diffuse large cell lymphoma [107]. An exception is thatin children with AIDS, <strong>the</strong> infiltrate more closely resemblesMALT lymphoma, <strong>and</strong> monoclonality may be demonstrated.5.7.5 Sclerosing PolycysticSialadenopathy(Sclerosing Polycystic Adenosis)This is a benign pseudoneoplastic condition <strong>of</strong> majorsalivary gl<strong>and</strong>s [11, 84, 200], said to be analogous to fibrocysticdisease <strong>of</strong> <strong>the</strong> breast [58]. It affects mainly femaleswith a mean age <strong>of</strong> 28 years (range 12 to 63). Mostcases have been described as slow-growing masses in <strong>the</strong>parotid gl<strong>and</strong>, with a single example <strong>of</strong> subm<strong>and</strong>ibulargl<strong>and</strong> involvement. The excised gl<strong>and</strong> is largely replaced


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 137by multiple discrete, firm, rubbery nodules. Microscopicexamination shows a well-circumscribed, unencapsulatedmass composed <strong>of</strong> a lobular arrangement <strong>of</strong> proliferatingducts <strong>and</strong> acini with cystic ducts containingviscous secretion <strong>and</strong>, on occasions, aggregates <strong>of</strong> foamymacrophages. There is <strong>of</strong>ten intraluminal epi<strong>the</strong>lialproliferation occasionally with a cribriform pattern <strong>and</strong><strong>the</strong>se may contain small droplets <strong>of</strong> basement membranematerial. The lining comprises a spectrum <strong>of</strong> apocrine,mucous, squamous cells <strong>and</strong> ballooned sebaceous-likecells, although true goblet cells are not seen. Some cellscontain prominent large, intensely eosinophilic cytoplasmicgranules <strong>of</strong> varying sizes, representing aberrantzymogen granules (Fig. 5.5). On occasion <strong>the</strong>re is nuclearpleomorphism, even suggesting dysplasia [200], but<strong>the</strong>re is no significant mitotic activity <strong>and</strong> no malignantcases have been described. Flattened myoepi<strong>the</strong>lial cellsare present around ductal <strong>and</strong> acinar structures, <strong>and</strong><strong>the</strong>re is periductal sclerosis <strong>and</strong> intense hyaline sclerosis<strong>of</strong> <strong>the</strong> surrounding s<strong>of</strong>t tissue. Sometimes, a patchylymphocytic infiltrate is noted. About one-third <strong>of</strong> casesrecur, but none has metastasised.5.7.6 O<strong>the</strong>r CystsO<strong>the</strong>r salivary cysts include dermoids [141], <strong>and</strong> a variety<strong>of</strong> epi<strong>the</strong>lial <strong>and</strong> non-epi<strong>the</strong>lial cysts including parasites<strong>and</strong> gas cysts in glass blowers [163]. Keratocystomais a rare, recently described, benign parotid tumourcharacterised by multicystic keratin-filled spaces linedwith stratified squamous epi<strong>the</strong>lium with no atypicalfeatures [150].5.8 Benign TumoursThere are various classifications. The revised WHOclassification (Table 5.1) [171] has <strong>the</strong> merit <strong>of</strong> being easilyapplicable in practice [181].5.8.1 Pleomorphic AdenomaFig. 5.6. Pleomorphic adenoma spectrum. Reproduced with permissionfrom Zarbo et al. [236]ICD-O:8940/0Most authors accept that <strong>the</strong>re is a spectrum <strong>of</strong> benignsalivary adenomas, including pleomorphic adenoma.Benign myoepi<strong>the</strong>lioma, which is composed almostentirely <strong>of</strong> myoepi<strong>the</strong>lial cells represents one end <strong>of</strong> <strong>the</strong>spectrum, whereas basal cell adenoma <strong>and</strong> canalicularadenoma are at <strong>the</strong> o<strong>the</strong>r end [183, 236, 237]. The particularmorphology <strong>of</strong> any particular tumour reflects <strong>the</strong>different proportions <strong>of</strong> <strong>the</strong> constituent cells (Fig. 5.6).Pleomorphic adenoma (PA) is <strong>the</strong> most common tumour<strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. Although most <strong>of</strong>ten foundin young to middle-aged women, <strong>the</strong>y can occur in ei<strong>the</strong>rsex <strong>and</strong> at any age. Up to 80% occur in <strong>the</strong> superficiallobe <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>, <strong>and</strong> it typically presentsas a painless swelling. When <strong>the</strong> deep lobe is involved,it <strong>of</strong>ten manifests as an intraoral parapharyngeal mass.Approximately 5% <strong>of</strong> PAs occur in <strong>the</strong> subm<strong>and</strong>ibulargl<strong>and</strong>, 0.1% in <strong>the</strong> sublingual gl<strong>and</strong> <strong>and</strong> about 10% inminor salivary gl<strong>and</strong>s [30, 228]. Similar tumours arisein extrasalivary locations including bronchi, ear, lacrimalgl<strong>and</strong>, breast <strong>and</strong> skin.Macroscopically, PAs are usually well-circumscribedmasses <strong>of</strong> 20–40 mm. The cut surface is usually white,<strong>and</strong> grey glistening areas are commonly seen.Histologically, PA was defined by <strong>the</strong> revised WHOClassification <strong>of</strong> 1991 as “a tumour [<strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong>s] <strong>of</strong> variable capsulation characterised microscopicallyby architectural ra<strong>the</strong>r than cellular pleomorphism”[171], i.e. <strong>the</strong> pleomorphism refers to <strong>the</strong> variety<strong>of</strong> histological patterns, not <strong>the</strong> cytology.The pattern varies from case to case, <strong>and</strong> also fromarea to area within any individual tumour. All are composed<strong>of</strong> a mixture <strong>of</strong> ductal epi<strong>the</strong>lial cells, basal <strong>and</strong>myoepi<strong>the</strong>lial cells <strong>and</strong> variable amounts <strong>of</strong> stroma,both hyaline <strong>and</strong> chondromyxoid. Attempts have beenmade to subclassify PA based on <strong>the</strong> proportions <strong>of</strong> celltypes <strong>and</strong> stroma [176], but because <strong>of</strong> <strong>the</strong> variation inany tumour, this is difficult <strong>and</strong> probably has no prognosticvalue.Ducts are lined with flat, cuboidal or columnar epi<strong>the</strong>lialcells, with little or no atypia. The ducts are usuallysmall tubules, but can be cystically dilated <strong>and</strong>also arranged in a cribriform pattern, resembling adenoidcystic carcinoma, but mitotic figures are rare <strong>and</strong><strong>the</strong> proliferation index low (see Sect. 5.9.3). Squamousmetaplasia with or without keratinisation is seen in upto 25% <strong>of</strong> PAs [65]. If associated with mucinous metaplasia,it may resemble mucoepidermoid carcinoma(Figs. 5.7, 5.8).Myoepi<strong>the</strong>lial cells are arranged in sheets, smallerisl<strong>and</strong>s <strong>and</strong> trabeculae, <strong>and</strong> also surround epi<strong>the</strong>-


138 S. Di Palma · R.H.W. Simpson · A. Skalova · I. LeivoTable 5.1. Revised WHO histological classification <strong>of</strong> salivary gl<strong>and</strong> tumours [171]5AdenomaDuctal papillomaCystadenomaCarcinomasNon-epi<strong>the</strong>lial tumoursMalignant lymphomasSecondary tumoursUnclassified tumoursEntities not included in <strong>the</strong> classification,but described or better characterised since1991 [8a]Pleomorphic adenomaMyoepi<strong>the</strong>lioma (myoepi<strong>the</strong>lial adenoma)Basal cell adenomaWarthin’s tumour (adenolymphoma)Oncocytoma (oncocytic adenoma)Canalicular adenomaSebaceous adenomaInverted ductal papillomaIntraductal papillomaSialadenoma papilliferumPapillary cystadenomaMucinous cystadenomaAcinic cell carcinomaMucoepidermoid carcinomaAdenoid cystic carcinomaPolymorphous low-grade adenocarcinoma (terminal duct adenocarcinoma)Epi<strong>the</strong>lial-myoepi<strong>the</strong>lial carcinomaBasal cell adenocarcinomaSebaceous carcinomaPapillary cystadenocarcinomaMucinous adenocarcinomaOncocytic carcinomaSalivary duct carcinomaAdenocarcinoma (not o<strong>the</strong>rwise specified)Malignant myoepi<strong>the</strong>lioma (myoepi<strong>the</strong>lial carcinoma)Carcinoma in pleomorphic adenomaSquamous cell carcinomaSmall cell carcinomaUndifferentiated carcinomaO<strong>the</strong>r carcinomasSialoblastomaHyalinising clear cell carcinomaCribriform adenocarcinoma <strong>of</strong> <strong>the</strong> tongueEndodermal sinus tumour <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>slium-lined spaces. As in benign myoepi<strong>the</strong>lioma (seeSect. 5.8.2), neoplastic myoepi<strong>the</strong>lial cells may take severalforms – epi<strong>the</strong>lioid, spindle, plasmacytoid, clear <strong>and</strong>oncocytic, as well as transitional forms with features <strong>of</strong>two or more <strong>of</strong> <strong>the</strong>se types (Fig. 5.9).The stroma varies in amount <strong>and</strong> is ei<strong>the</strong>r dense eosinophilichyaline material or chondromyxoid tissue.The former is composed <strong>of</strong> basement membrane material<strong>and</strong> stains with PAS diastase <strong>and</strong> collagen type IV;<strong>the</strong> chondromyxoid material only rarely resembles truecartilage <strong>and</strong> is Alcian blue-positive (Fig. 5.10). Calcification<strong>and</strong> bone formation can occur in long st<strong>and</strong>ingtumours. Occasionally, collagenous spherules <strong>and</strong> crystalloidsare seen, particularly in tumours rich in myoepi<strong>the</strong>lialcells <strong>of</strong> <strong>the</strong> plasmacytoid type (Fig. 5.11) [197].Nuclear atypia is not common, but can be seen in tumourswhere epi<strong>the</strong>lial or myoepi<strong>the</strong>lial cells display oncocyticfeatures [65]. Occasional myoepi<strong>the</strong>lial cell nucleiare enlarged <strong>and</strong> bizarre, somewhat analogous to“ancient” change in schwannomas. Mitotic figures aregenerally sparse, but can occur as part <strong>of</strong> <strong>the</strong> repair processafter FNA. Such tumours with <strong>the</strong>se atypical featuresshould be sampled thoroughly to exclude true intracapsularcarcinoma.Similarly, areas <strong>of</strong> necrosis or haemorrhage may followsurgical manipulation, FNA or o<strong>the</strong>r trauma, <strong>and</strong>


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 139Fig. 5.7. Pleomorphic adenoma: myoepi<strong>the</strong>lial cells with an epi<strong>the</strong>lioidcytomorphology. These cells may also be spindle-shaped,plasmacytoid (hyaline) or have clear cytoplasm. Note also a smallduct <strong>and</strong> a focus <strong>of</strong> squamous metaplasia. Keratinising squamousmetaplasia is seen in up to a quarter <strong>of</strong> pleomorphic adenomasFig. 5.10. Pleomorphic adenoma: chondromyxoid stroma containingisolated small <strong>and</strong> small aggregates <strong>of</strong> myoepi<strong>the</strong>lialcellsFig. 5.8. Pleomorphic adenoma with squamous <strong>and</strong> focal mucinousmetaplasia resembling mucoepidermoid carcinomaFig. 5.11. Collagenous spherules can be seen in some benign myoepi<strong>the</strong>liomas<strong>and</strong> myoepi<strong>the</strong>lium-rich pleomorphic adenomasFig. 5.9. Pleomorphic adenoma: myoepi<strong>the</strong>lial cells showing anepi<strong>the</strong>lioid <strong>and</strong> plasmocytoid appearanceFig. 5.12. Vascular “invasion” is a rare finding in benign pleomorphicadenoma, due to displacement <strong>of</strong> neoplastic cells intovascular spaces. It is not indicative <strong>of</strong> malignancy


140 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5Fig. 5.13. Recurrent pleomorphic adenoma. Multiple <strong>and</strong> <strong>of</strong>tenwell-separated tumour nodules <strong>of</strong> different sizes are seen in <strong>the</strong>periparotid s<strong>of</strong>t tissueFig. 5.14. Benign myoepi<strong>the</strong>lioma composed <strong>of</strong> plasmocytoid(hyaline) <strong>and</strong> epi<strong>the</strong>lioid cells with areas <strong>of</strong> myxoid stroma. Plasmocytoidcells have eccentric nuclei <strong>and</strong> dense eosinophilic cytoplasm<strong>the</strong>se neoplasms should also be sampled thoroughly. Tumourcells in lymphatics (“vascular invasion”) are occasionallyseen in benign PAs, but this does not necessarilyindicate malignancy (Fig. 5.12) [3]. None <strong>of</strong> <strong>the</strong> reportedcases were followed by metastases.Pleomorphic adenomas are <strong>of</strong>ten completely or partlysurrounded by a fibrous capsule <strong>of</strong> variable thickness,but it can be absent, especially in tumours <strong>of</strong> <strong>the</strong> minorgl<strong>and</strong>s. Neoplastic elements may extend into <strong>and</strong> eventhrough <strong>the</strong> capsule in <strong>the</strong> form <strong>of</strong> microscopic pseudopodiaor apparent satellite nodules.They may be <strong>the</strong> cause <strong>of</strong> future recurrence after apparentsurgical removal [97], <strong>and</strong> <strong>the</strong>ir presence shouldbe noted in <strong>the</strong> surgical pathology report. Special stains<strong>and</strong> immunohistochemistry are not necessary for <strong>the</strong>diagnosis in most cases, but can be used to identify <strong>the</strong>different cell types <strong>and</strong> also early malignant change (seeSect. 5.9.11).Recurrent PA occurs after incomplete surgical excision<strong>and</strong> is usually composed <strong>of</strong> multiple nodules completelyseparate from each o<strong>the</strong>r. In <strong>the</strong> first recurrence<strong>the</strong> nodules are usually seen within salivary gl<strong>and</strong> tissue,but in fur<strong>the</strong>r recurrences tumours are found in <strong>the</strong>s<strong>of</strong>t tissue <strong>of</strong> <strong>the</strong> surgical bed (Fig. 5.13). Histologically,<strong>the</strong> nodules show similar features to ordinary PA, <strong>and</strong>in particular <strong>the</strong>y lack any cytological atypia. In spite<strong>of</strong> this, confluent nodules <strong>of</strong> recurrent PA can still kill<strong>the</strong> patient. As discussed later (see Sect. 5.9.11) multiplyrecurrent PAs may rarely metastasise to distant sites,<strong>and</strong> in addition are more prone to developing malignantchanges.5.8.1.1 Salivary Gl<strong>and</strong> Anlage Tumour(“ Congenital PleomorphicAdenoma”)ICD-O:8940/0This is a rare, probably hamartomatous lesion in <strong>the</strong> nasopharynx<strong>of</strong> neonates [45]. Although potentially fataldue to its location, prognosis after surgery is good. It wasnot included in <strong>the</strong> 1991 WHO classification [171]. Themicroscopic features are a biphasic pattern <strong>of</strong> squamousnests <strong>and</strong> duct-like structures at <strong>the</strong> periphery, merginginto solid, predominantly mesenchymal nodules, possibly<strong>of</strong> myoepi<strong>the</strong>lial origin. Occasionally, <strong>the</strong>re is necrosis<strong>and</strong> cyst formation [136].5.8.2 Benign Myoepi<strong>the</strong>liomaICD-O:8982/0Myoepi<strong>the</strong>lial cells are found in several salivary gl<strong>and</strong>neoplasms (Table 5.2). Benign myoepi<strong>the</strong>lioma wasfirst described in 1943 [179], <strong>and</strong> was included in<strong>the</strong> 1991 revised WHO classification [171]. It can bedefined as a tumour composed totally, or almost totally,<strong>of</strong> myoepi<strong>the</strong>lial cells. Whe<strong>the</strong>r or not it is trulya separate biological entity is debatable, but mostcommentators believe that it represents one end <strong>of</strong> aspectrum that also includes pleomorphic <strong>and</strong> at leastsome basal cell adenomas. Never<strong>the</strong>less, myoepi<strong>the</strong>liomadisplays particular microscopic features thatpose specific practical problems in <strong>the</strong> identification<strong>and</strong> differential diagnosis, <strong>and</strong> on this basis it can beaccepted as a separate diagnostic category [188, 189].Most cases present as a well-circumscribed mass, usu-


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 141Table 5.2. Salivary tumours with myoepi<strong>the</strong>lial cell participation. Adapted from <strong>the</strong> WHO classification [171]BenignMalignantPleomorphic adenomaMyoepi<strong>the</strong>liomaBasal cell adenoma (some)Adenoid cystic carcinomaPolymorphous low-grade adenocarcinomaEpi<strong>the</strong>lial-myoepi<strong>the</strong>lial carcinomaMalignant myoepi<strong>the</strong>lioma (myoepi<strong>the</strong>lial carcinoma)Carcinoma ex pleomorphic adenoma (some)ally 10–50 mm in diameter, in ei<strong>the</strong>r major or minorsalivary gl<strong>and</strong>s. Microscopically, <strong>the</strong>re are several typicalappearances, reflecting <strong>the</strong> different forms thatneoplastic myoepi<strong>the</strong>lial cells can take. Solid, myxoid<strong>and</strong> reticular growth patterns may be seen, <strong>and</strong> <strong>the</strong>component cells may be spindle-shaped, plasmacytoid(hyaline), clear, epi<strong>the</strong>lioid or oncocytic. Manytumours show more than one growth pattern or celltype, but myoepi<strong>the</strong>liomas <strong>of</strong> <strong>the</strong> minor gl<strong>and</strong>s aremore <strong>of</strong>ten composed <strong>of</strong> plasmacytoid cells, <strong>and</strong> those<strong>of</strong> <strong>the</strong> parotid spindle cells [189]. Although most authorsaccept <strong>the</strong> plasmacytoid cells as myoepi<strong>the</strong>lial,it has recently been suggested that <strong>the</strong>se cells originatefrom luminal <strong>and</strong> not from myoepi<strong>the</strong>lial cells [157],<strong>and</strong> thus <strong>the</strong> tumours should possibly be reclassifiedas plasmacytoid adenomas [157]. The clear cell variantcan occur in both major <strong>and</strong> minor gl<strong>and</strong>s [182], butis relatively rare [43]. Unlike <strong>the</strong>ir malignant counterpart[52] (see Sect. 5.9.8), benign myoepi<strong>the</strong>liomas donot usually show invasiveness, necrosis, cytologicalpleomorphism, or more than an isolated mitotic figure.The stroma is usually scanty, fibrous or myxoid,<strong>and</strong> it may occasionally contain chondroid material ormature fat cells [203]. Extracellular collagenous crystalloidsare seen in 10–20% <strong>of</strong> plasmacytoid cell-typemyoepi<strong>the</strong>liomas, (as well as sometimes in myoepi<strong>the</strong>lial-richPAs); <strong>the</strong>se structures are about 50–100 min diameter <strong>and</strong> consist <strong>of</strong> radially-arranged needleshapedfibres composed <strong>of</strong> collagen types I <strong>and</strong> III,which stain red with <strong>the</strong> van Gieson method [197].Scanty small ducts may be present (usually less than10% <strong>of</strong> <strong>the</strong> tumour tissue) in o<strong>the</strong>rwise typical myoepi<strong>the</strong>liomas(Fig. 5.14) [43]. Immunohistochemically,almost all tumours express S-100 protein, as wellas some cytokeratins, especially subtype 14. Alphasmooth muscle actin positivity is seen to some degreein most spindle cell myoepi<strong>the</strong>liomas, but only occasionallyin <strong>the</strong> plasmacytoid cell type [189]. Stainingfor calponin, smooth muscle myosin heavy chain(SMMHC) <strong>and</strong> CD10 is inconsistent in myoepi<strong>the</strong>lialcells. The nuclear transcription factor p63 is positivein most benign myoepi<strong>the</strong>liomas [166]. Electron microscopicstudies have also confirmed both epi<strong>the</strong>lialFig. 5.15. Basal cell adenoma. The tumour is arranged in nests, isl<strong>and</strong>s<strong>and</strong> trabeculae or basal cells without cytological abnormality.Ductal differentiation is also noted<strong>and</strong> smooth muscle differentiation [170], although focaldensities in my<strong>of</strong>ilaments are not usually found[43]. The behaviour <strong>of</strong> myoepi<strong>the</strong>lioma is similar tothat <strong>of</strong> pleomorphic adenoma, <strong>and</strong> complete excisionshould be curative. Nei<strong>the</strong>r growth pattern nor celltype appears to carry prognostic significance. Malignantchange in a benign lesion has been described [2],but too little information is available about <strong>the</strong> percentage<strong>of</strong> cases involved. However, it is reasonable topostulate that it is probably not very different fromthat <strong>of</strong> pleomorphic adenoma.5.8.3 Basal Cell AdenomaICD-O:8147/0Most tumours previously described as monomorphicadenoma are now termed basal cell adenoma (BCA).The revised WHO [171] classification recognises fourhistopathological subtypes – solid, tubular, trabecular<strong>and</strong> membranous – but it is likely that, in reality, <strong>the</strong>reare only two separate biological entities [16] – membranous<strong>and</strong> non-membranous (Figs 5.15, 5.16).


142 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5Fig. 5.16. Membranous basal cell adenoma: jigsaw like pattern:multiple epi<strong>the</strong>lial isl<strong>and</strong>s surrounded by large amounts <strong>of</strong> basalmembrane-like material. The latter is also present within <strong>the</strong> cytoplasm<strong>of</strong> some <strong>of</strong> <strong>the</strong> small dark hyperchromatic basal cells. Thereis little cellular pleomorphismNon-membranous BCAs have an equal sex incidence<strong>and</strong> arise mostly in <strong>the</strong> major gl<strong>and</strong>s. They probablyrepresent part <strong>of</strong> <strong>the</strong> spectrum <strong>of</strong> myoepi<strong>the</strong>lioma<strong>and</strong> pleomorphic adenoma [70, 237]. The tumoursare ovoid, well-circumscribed masses in which isl<strong>and</strong>s,nests <strong>and</strong> trabeculae <strong>of</strong> basaloid cells are eachsurrounded by a distinct thin PAS-positive basementmembrane. The component cells may take two forms –small with scanty cytoplasm <strong>and</strong> a round, dark nucleus,<strong>and</strong> larger with amphophilic or eosinophilic cytoplasm<strong>and</strong> an ovoid paler staining nucleus. These twotypes are intermixed, but <strong>the</strong> smaller cells tend to bearranged around <strong>the</strong> periphery <strong>of</strong> <strong>the</strong> nests <strong>and</strong> trabeculae,giving <strong>the</strong> appearance <strong>of</strong> palisading. Ductal differentiationmay or may not be apparent, but can behighlighted by EMA. There is little pleomorphism <strong>and</strong>mitotic figures are rare. The stroma varies in amount<strong>and</strong> cellularity, but S-100 protein-positive spindle cellsmay be numerous. S-100 positive cells are also presentwithin <strong>the</strong> isl<strong>and</strong>s <strong>of</strong> epi<strong>the</strong>lial cells, which reactstrongly with cytokeratins [220].Membranous BCA (dermal analogue tumour) occurspredominantly in men, <strong>and</strong> can be multicentric.Most arise in <strong>the</strong> major gl<strong>and</strong>s, including within intraparotidlymph nodes [128]. Microscopically, <strong>the</strong>yare not encapsulated <strong>and</strong> appear multinodular, <strong>of</strong>tenwith a jigsaw-like pattern. The most characteristicfeature is <strong>the</strong> deposition <strong>of</strong> large amounts <strong>of</strong> hyalinebasement membrane material, which is brightly eosinophilic<strong>and</strong> PAS-positive. It surrounds <strong>the</strong> epi<strong>the</strong>lialcell isl<strong>and</strong>s <strong>and</strong> blood vessels, <strong>and</strong> is present within<strong>the</strong> isl<strong>and</strong>s as small droplets. There is little pleomorphismor mitotic activity. In about 40% <strong>of</strong> cases, <strong>the</strong>salivary adenoma is associated with synchronous <strong>and</strong><strong>of</strong>ten multiple skin appendage tumours <strong>of</strong> sweat gl<strong>and</strong>or hair follicle origin, usually cylindromas or eccrinespiradenomas.The most important differential diagnosis <strong>of</strong> all types<strong>of</strong> BCA is adenoid cystic carcinoma. Useful pointers toadenoma include lack <strong>of</strong> invasiveness <strong>and</strong> cytologicalpleomorphism, low mitotic <strong>and</strong> proliferative activity,<strong>and</strong> whorled eddies <strong>of</strong> epi<strong>the</strong>lial cells. S-100 protein positivity<strong>of</strong> spindled stromal cells may help, as this doesnot occur in adenoid cystic carcinoma [70]. BCA closelyresembles basal cell adenocarcinoma, which may lackcytological pleomorphism <strong>and</strong> mitotic figures, <strong>the</strong> diagnosis<strong>the</strong>n depending principally on <strong>the</strong> presence <strong>of</strong> genuineinvasion (see Sect. 5.9.7).The recurrence rate for non-membranous BCA isextremely low (0 out <strong>of</strong> 102 patients in one series) [16],<strong>and</strong> local excision with clear margins is sufficient treatment.There is a low rate <strong>of</strong> malignant transformation(about 4%) into basal cell adenocarcinoma [127]. In contrast,up to 24% <strong>of</strong> membranous BCAs recur after surgery[16], probably reflecting multicentricity <strong>and</strong>, in addition,malignancy (also as basal cell adenocarcinoma)develops in 28% [127]. Surgery for this subtype needs tobe more extensive [16, 119, 130].5.8.4 Warthin’s TumourICD-O:8561/0Warthin’s tumour (WT; adenolymphoma) is <strong>the</strong> secondcommonest neoplasm <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>, <strong>and</strong> is <strong>the</strong>easiest salivary tumour to diagnose by microscopy [66].It arises almost exclusively in <strong>the</strong> parotid gl<strong>and</strong> (usually<strong>the</strong> tail) <strong>and</strong> occasionally in periparotid lymph nodes.The mean age at diagnosis is 62 years (range 29–88),<strong>and</strong> WT is uncommon in blacks. Previously, <strong>the</strong>re was amarked male predominance (as much as 26:1), but now<strong>the</strong>re is an almost equal sex distribution. It is eight timesmore frequent in heavy smokers. WT is multicentric in12% <strong>of</strong> patients, <strong>and</strong> bilateral in 6%.There are two <strong>the</strong>ories <strong>of</strong> its histogenesis [62] – a trueepi<strong>the</strong>lial neoplasm that attracts a heavy lymphoid reaction,or alternatively a non-neoplastic lesion arisingfrom ectopic salivary inclusions in intraparotid lymphnodes. The latter <strong>the</strong>ory is supported by a molecular geneticstudy using HUMARA analysis, which has shownthat WT is not a clonal process [100].Pathological examination shows a well-circumscribedoval mass, composed <strong>of</strong> slits or cystic spaceswith papillary infoldings lined with two layers <strong>of</strong>oncocytic epi<strong>the</strong>lium; <strong>the</strong> inner cells are columnarwith nuclear palisading, deep to which are flattenedor cuboidal basal cells (Fig. 5.17). Occasional mucous<strong>and</strong> squamous cells may be seen. The stroma comprisesusually plentiful lymphoid tissue with germinalcentre formation. Special stains <strong>and</strong> immunohistochemistryhave little to <strong>of</strong>fer in practice; myoepi-


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 143Fig. 5.17. Warthin’s tumour. Cystic <strong>and</strong> slit-like spaces with papillaryinfolding lined with oncocytic cells. Lymphoid tissue occupies<strong>the</strong> cores <strong>of</strong> most papillaeFig. 5.20. Canalicular adenoma <strong>of</strong> <strong>the</strong> upper lip. It is composed<strong>of</strong> bi-layered str<strong>and</strong>s <strong>of</strong> basal-like cells embedded in a loose oedematousstroma<strong>the</strong>lial markers are negative. Histological variants includea stroma-poor form, <strong>and</strong> metaplastic WT – in<strong>the</strong> latter, much <strong>of</strong> <strong>the</strong> original oncocytic epi<strong>the</strong>liumhas been replaced by squamous cells, <strong>and</strong> <strong>the</strong>re is extensivenecrosis, fibrosis, inflammation, <strong>and</strong> granulomaformation (Fig. 5.18) [66]. This not uncommonlesion follows trauma, particularly FNA [55], <strong>and</strong> canbe mistaken for squamous or mucoepidermoid carcinomas(see Sect. 5.5.5). WT generally has a good outcome,with recurrence rates <strong>of</strong> about 2%. Malignancyoccurs in less than 1% <strong>of</strong> cases, involving ei<strong>the</strong>r epi<strong>the</strong>lialor lymphoid elements leading to carcinomas orlymphomas [62].Fig. 5.18. Metaplastic (infarcted) Warthin’s tumour. There is extensivenecrosis. A surrounding thin rim <strong>of</strong> viable tissue showssquamous metaplasia5.8.5 OncocytomaICD-O:8290/0Oncocytic change in salivary tumours is common (seeSect. 5.6) [65]. Oncocytoma is a true benign neoplasmcomposed <strong>of</strong> oncocytes. It is rare <strong>and</strong> is <strong>of</strong>ten associatedwith MNOH (see Sect. 5.6) [158]. It comprises awell-demarcated mass <strong>of</strong> oncocytic cells (both light <strong>and</strong>dark) with a solid, trabecular, or tubular configuration(Fig. 5.19). There is a surrounding, usually incompletefibrous capsule, <strong>and</strong> only a little internal fibrous stroma.There is a rare clear cell variant [61].5.8.6 Canalicular AdenomaFig. 5.19. Oncocytoma. Light <strong>and</strong> dark oncocytic cells are arrangedin a solid, trabecular <strong>and</strong> tubular configurationICD-O:8149/0Canalicular adenoma also has a basaloid appearance.Its location is almost exclusively intraoral, particularlyaffecting <strong>the</strong> upper lip [119] <strong>and</strong> less <strong>of</strong>ten <strong>the</strong>palate. As a result, most tumours present when small


144 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5– rarely more than 20 mm in diameter. It has a characteristicmorphology <strong>of</strong> branching <strong>and</strong> interconnectingbi-layered str<strong>and</strong>s <strong>of</strong> darkly staining epi<strong>the</strong>lialcells set in a loose vascular stroma (Fig. 5.20). Thereis no pleomorphism or significant mitotic activity.The cells express cytokeratins <strong>and</strong> S-100 protein. Notinfrequently, <strong>the</strong>y are multifocal [39], <strong>and</strong> can thusmimic <strong>the</strong> true invasiveness <strong>of</strong> cribriform adenoidcystic carcinoma. The lack <strong>of</strong> destructiveness <strong>and</strong> <strong>the</strong>presence <strong>of</strong> blood vessels in <strong>the</strong> cribriform spaces aregood guides to canalicular adenoma, which is completelybenign. Occasional recurrences are a result <strong>of</strong>multifocality [90].5.8.7 Sebaceous AdenomaICD-O:8410/0This rare, encapsulated epi<strong>the</strong>lial tumour is composed<strong>of</strong> solid, variably shaped isl<strong>and</strong>s <strong>and</strong> cysts, both showingfocal sebaceous differentiation with squamous areas;<strong>the</strong>se are surrounded by a fibrous, hyalinised stroma.They do not recur after complete surgical excision [12,62].5.8.8 Sebaceous LymphadenomaICD-O:8410/0This lesion comprises irregular proliferating nests <strong>and</strong>isl<strong>and</strong>s <strong>of</strong> epi<strong>the</strong>lium including solid <strong>and</strong> gl<strong>and</strong>-like sebaceouselements, surrounded by a lymphoid stroma.It is possible that, like Warthin’s tumour, sebaceouslymphadenoma develops from salivary inclusions withinlymph nodes, <strong>and</strong> shows sebaceous ra<strong>the</strong>r than oncocyticmetaplasia [62, 171].5.8.9 Ductal PapillomaICD-O:8503/0There are three subtypes, all rare – inverted ductal papilloma(similar to <strong>the</strong> sinonasal tumour), intraductalpapilloma <strong>and</strong> sialadenoma papilliferum (similar toskin syringocystadenoma papilliferum) [62]. Intraductalpapilloma has a fibrovascular core lined with myoepi<strong>the</strong>lial<strong>and</strong> ductal cells <strong>and</strong> it is usually seen in a dilatedduct.5.8.10 CystadenomaFig. 5.21. Oncocytic (papillary) cystadenoma <strong>of</strong> <strong>the</strong> larynx. Cysticallydilated ducts are lined with oncocytic cellsICD-O:8440/0The revised WHO classification recognises two histopathologicalsubtypes –papillary cystadenoma (similarto lymphoid-poor Warthin’s tumour) <strong>and</strong> mucinouscystadenoma. Both are rare, benign tumourscharacterised by unicystic or multicystic growth patterns.The latter can be mistaken for mucinous malignancy,such as grade I mucoepidermoid carcinoma[62, 173]. Most cystadenomas are multilocular withindividual cystic spaces separated by limited amounts<strong>of</strong> intervening stroma [229]. The lumina <strong>of</strong>ten containeosinophilic material with scattered epi<strong>the</strong>lial, foamyor inflammatory cells. Rarely, psammoma bodies <strong>and</strong>crystalloids have been described within <strong>the</strong> luminalsecretion [199]. The lining epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong> cysticspaces is mostly columnar <strong>and</strong> cuboidal. Oncocytic,mucous <strong>and</strong> apocrine cells are sometimes present focallyor may predominate. An oncocytic variant <strong>of</strong>papillary cystadenoma is composed <strong>of</strong> oncocytes presentin unilayered or bilayered papillary structures(Fig. 5.21). Squamous epi<strong>the</strong>lium may be present, butrarely predominates. The tumours are unlikely to recur,but rare cases <strong>of</strong> mucinous cystadenoma with malignanttransformation have been described (Figs 5.22,5.23) [135].5.9 Malignant Epi<strong>the</strong>lialTumours5.9.1 Acinic Cell CarcinomaICD-O:8550/3Acinic cell carcinoma (AcCC) is defined as a malignantepi<strong>the</strong>lial neoplasm in which some <strong>of</strong> <strong>the</strong> neoplastic cellsdemonstrate serous acinar cell differentiation.It accounts for about 2–4% <strong>of</strong> salivary gl<strong>and</strong> tumours,<strong>and</strong> 12–17% <strong>of</strong> malignancies [62]. It is slightlycommoner in women <strong>and</strong> <strong>the</strong> mean age at presentationis 44 years, although AcCCs can affect children


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 145Fig. 5.22. Mucinous cystadenoma. Cysts are lined with mucussecretingcells without atypiaFig. 5.24. Acinic cell carcinoma solid variant. The cells showgranular cytoplasm <strong>and</strong> acinar differentiation similar to normalsalivary gl<strong>and</strong> aciniFig. 5.23. Mucinous cystadenoma with malignant transformation[136]. Cellular pleomorphism <strong>and</strong> signet ring cell appearanceFig. 5.25. Acinic cell carcinoma, papillary subtype: papillae arelined with intercalated duct-like cells, some containing microvesicles,o<strong>the</strong>rs showing a hobnail/clear cell appearance<strong>and</strong> centenarians. The parotid is involved in 92% <strong>of</strong>cases (3% bilateral), with only occasional examples in<strong>the</strong> subm<strong>and</strong>ibular or minor gl<strong>and</strong>s [62], or periparotidlymph nodes [161]. The typical clinical history is <strong>of</strong>a slowly enlarging mass (for as long as 40 years) sometimeswith pain <strong>and</strong> facial nerve weakness. Most tumoursare partly circumscribed, with a diameter <strong>of</strong> 10–30 mm, although some may reach 220 mm [4]. Microscopyshows one or more growth patterns – solid, microcystic,follicular <strong>and</strong> papillary-cystic. The follicularpattern resembles thyroid tissue, <strong>and</strong> any tumour in<strong>the</strong> parotid with a papillary-cystic architecture shouldbe considered to be an AcCC, until proven o<strong>the</strong>rwise.The cells in AcCC may take one <strong>of</strong> several forms – acinar(serous or blue dot), intercalated ductal (cuboidal,<strong>of</strong>ten lining small ducts), microvesicular, hob-nail orclear – <strong>the</strong> last is surprisingly rare, being seen in only6% <strong>of</strong> cases [62]. Several growth patterns <strong>and</strong> cell typesmay be seen in any individual tumour (Figs 5.24–5.26).Dedifferentiation towards a high-grade malignancyoccurs occasionally [51], <strong>and</strong> all surgical specimens <strong>of</strong>AcCC must be sampled adequately. A lymphoid infiltrateis found in about 30% <strong>of</strong> cases, but is only <strong>of</strong> clinicalsignificance (having a good prognosis) when <strong>the</strong>tumour is a well-circumscribed nodule with a micr<strong>of</strong>olliculararchitecture (Fig. 5.27) [133].The most useful special stain in AcCC is PASD,which highlights cytoplasmic zymogen granules(Fig. 5.28). Immunohistochemistry is <strong>of</strong> limited value– positivity is seen with cytokeratin, amylase, CEA<strong>and</strong> in 10% S-100 protein [221]. O<strong>the</strong>r myoepi<strong>the</strong>lialmarkers are negative. It has been suggested that bonemorphogenetic protein-6 (BMP-6) may be useful, butthis marker is not yet widely available [92]. Electron


146 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5Fig. 5.26. Follicular variant <strong>of</strong> acinic cell carcinoma: <strong>the</strong> tumouris composed <strong>of</strong> follicle-like spaces <strong>of</strong> varying sizes lined with cuboidalintercalated duct-type cellsFig. 5.28. Acinic cell carcinoma. PAS-D emphasises coarse zymogengranules in <strong>the</strong> cytoplasm <strong>of</strong> <strong>the</strong> tumour cellsthat Ki-67 (MIB1) is an independent prognostic indicator[94, 198]. Skálová et al. found that tumours witha proliferation index


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 147Fig. 5.29. Mucoepidermoid carcinoma: clear, intermediate <strong>and</strong>mucus-secreting cellsFig. 5.31. High-grade mucoepidermoid carcinoma: “epidermoid”cells arranged in a solid pattern also show nuclear pleomorphism.Mucus-secreting cells may be scarcepredict outcome to some degree, <strong>and</strong> MECs should begiven one <strong>of</strong> three microscopic grades, based on <strong>the</strong> extent<strong>of</strong> <strong>the</strong> cystic component, neural invasion, necrosis,cytological pleomorphism <strong>and</strong> mitotic activity. Thisassessment has considerable prognostic significance,with death rates due to disease <strong>of</strong> 3.3, 9.7 <strong>and</strong> 46.3%for grades 1, 2 <strong>and</strong> 3 respectively [62]. Recently, a newgrading system has been proposed, but it is still underevaluation [22]. Assessment <strong>of</strong> <strong>the</strong> MIB1 proliferationindex has also been shown to be <strong>of</strong> value [196].5.9.3 Adenoid Cystic CarcinomaFig. 5.30. Low-grade mucoepidermoid carcinoma: typical cystic<strong>and</strong> solid patternance (Figs. 5.29–5.31). In fact, keratinisation is veryrare in MEC, <strong>and</strong> indeed is much commoner as part<strong>of</strong> squamous metaplasia in pleomorphic adenoma ormalignant myoepi<strong>the</strong>lioma, <strong>and</strong> in metastatic squamouscarcinoma from <strong>the</strong> skin or upper aerodigestivetract. Epidermoid cells may be sparse in MECs, <strong>and</strong>high molecular weight cytokeratin stains (e.g. LP34)<strong>and</strong> p63 can help identify <strong>the</strong>m. Intermediate cells aresmall with dark-staining nuclei <strong>and</strong> <strong>the</strong>y <strong>of</strong>ten form<strong>the</strong> stratified lining <strong>of</strong> cysts beneath <strong>the</strong> mucous cells.Clear cell change may be seen in ei<strong>the</strong>r <strong>the</strong> squamousor intermediate cells <strong>and</strong> MEC may take <strong>the</strong> form <strong>of</strong> aclear cell carcinoma [182]. Similarly, oncocytes can beplentiful [109]. All MECs are malignant with a metastaticpotential, regardless <strong>of</strong> <strong>the</strong>ir microscopic appearance.Never<strong>the</strong>less, histological features can be used toICD-O:8200/3Adenoid cystic carcinoma (AdCC) is a malignant tumourwith no particular age or sex predilection. It canoccur in any gl<strong>and</strong>, but most <strong>of</strong>ten in <strong>the</strong> subm<strong>and</strong>ibularor minor salivary gl<strong>and</strong>s, particularly <strong>the</strong> palate. However,in spite <strong>of</strong> <strong>of</strong>ten apparently slow growth, outcomeover <strong>the</strong> long term is poor. AdCC is an extensively infiltrativetumour with characteristic perineural invasion,<strong>and</strong> this is partly responsible for <strong>the</strong> clinical presentation<strong>of</strong> late, but repeated local recurrences. Unlike o<strong>the</strong>rsalivary gl<strong>and</strong> malignancies, when AdCC metastasises,it tends to involve distant organs (lung, bone) ra<strong>the</strong>rthan local lymph nodes [117].Histologically, AdCC is a generally solid tumour inwhich <strong>the</strong> cribriform pattern is easily recognised on microscopy,but tubular <strong>and</strong> solid structures can also bepresent. The commonest growth patterns are:Cribriform: this is <strong>the</strong> most characteristic microscopicfeature, dominated by multiple cribriform structures,composed <strong>of</strong> epi<strong>the</strong>lial <strong>and</strong> basal/myoepi<strong>the</strong>lial cells.The nuclei are usually dark, hyperchromatic <strong>and</strong> angulated.Mitotic figures are easy to find <strong>and</strong> may be abun-


148 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5Fig. 5.32. Adenoid cystic carcinoma, cribriform variant: multiplecribriform spaces composed <strong>of</strong> basaloid cells, with hyalinised materialsurrounded by small hyperchromatic cellsFig. 5.34. Adenoid cystic carcinoma, solid variant. This is composed<strong>of</strong> multiple solid nodules, some displaying central comedolikenecrosis. The tumour can be seen to infiltrate boneFig. 5.33. Adenoid cystic carcinoma, cribriform variant. Diffusehyalinisation with compression <strong>of</strong> tumour cells. Nuclear pleomorphismmay be difficult to appreciate, leading to a false diagnosis <strong>of</strong>pleomorphic adenomaFig. 5.35. Adenoid cystic carcinoma, solid variant. Tumour isl<strong>and</strong>scontain small ducts lined with a layer <strong>of</strong> epi<strong>the</strong>lial cells. In<strong>the</strong> absence <strong>of</strong> characteristic cribriform structures, <strong>the</strong> latter featureis diagnosticdant; <strong>the</strong> MIB1 proliferation index exceeds 10% [201].The contents <strong>of</strong> <strong>the</strong> spaces can be loose <strong>and</strong> basophilicor dense <strong>and</strong> eosinophilic. Hyalinisation is commonin adenoid cystic carcinoma <strong>and</strong> may be extreme. Inthose cases with excessive deposition <strong>of</strong> hyalinised material,<strong>the</strong> spaces are distended with loss <strong>of</strong> <strong>the</strong> cribriformpattern. Tumour cells may be sparse <strong>and</strong> bl<strong>and</strong>,<strong>and</strong> thus <strong>the</strong> lesions may mimic a pleomorphic adenoma(Figs. 5.32, 5.33).Tubular: this is composed <strong>of</strong> small tubules lined withone or two cell types, luminal <strong>and</strong> abluminal withoutsignificant cytological atypia. Because <strong>of</strong> this bl<strong>and</strong> cytologicalappearance it may be mistaken for basal celladenoma, except for <strong>the</strong> presence <strong>of</strong> infiltration.Solid (basaloid): this is dominated by large solid sheets<strong>of</strong> tumour cells, sometimes with comedo-like centralnecrosis. Within <strong>the</strong> solid masses <strong>of</strong> tumour cells, <strong>the</strong>reare small duct-like spaces surrounded by a definite layer<strong>of</strong> epi<strong>the</strong>lial cells (Figs. 5.34, 5.35). This latter findingdistinguishes solid variant AdCC from (relatively lowgrade)basal cell adenocarcinoma <strong>and</strong> <strong>the</strong> aggressive basaloidsquamous cell carcinoma, which in addition <strong>of</strong>tenshows intraepi<strong>the</strong>lial dysplastic changes.A rare finding in all types <strong>of</strong> AdCC is squamousmetaplasia, ei<strong>the</strong>r as single cells or with keratin pearlformation [62].A system <strong>of</strong> three grades based on <strong>the</strong> presence <strong>of</strong>tubular, cribriform <strong>and</strong> solid pattern [171] has shown


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 149that outcome is better in tubular ACC, while <strong>the</strong> worstprognosis is seen in solid AdCC. Never<strong>the</strong>less, clinicalstage appears to be a better predictor than grade[210].Ano<strong>the</strong>r unfavourable feature <strong>of</strong> AdCC is <strong>the</strong> frequentinvolvement <strong>of</strong> resection margins in <strong>the</strong> surgicalspecimen, particularly as <strong>the</strong> result <strong>of</strong> extensive perineuralinfiltration. As complete excision <strong>of</strong> AdCC is difficult,patients <strong>of</strong>ten require postoperative radio<strong>the</strong>rapy.A most important histological differential diagnosisis between AdCC <strong>and</strong> polymorphous low-grade adenocarcinoma(see Sect. 5.9.4).5.9.4 PolymorphousLow-Grade AdenocarcinomaFig. 5.36. Polymorphous low-grade adenocarcinoma. Perineuralinfiltration. Tumour cells show bl<strong>and</strong> cytonuclear abnormalityFig. 5.37. Polymorphous low-grade adenocarcinoma. Indian filingappearance resembling lobular carcinoma <strong>of</strong> <strong>the</strong> breastICD-O:8525/3Polymorphous low-grade adenocarcinoma (PLGA) isalso known as terminal duct or lobular carcinoma. It ismore frequent in women, <strong>and</strong> <strong>the</strong> average age at presentationis 59 years (range 21–94) [62]. Most cases arise inintra-oral minor salivary gl<strong>and</strong>s, particularly <strong>the</strong> palate,with only rare examples in <strong>the</strong> parotid, sometimes developinginto a pleomorphic adenoma [223]. The characteristichistological picture <strong>of</strong> PLGA is an infiltratingtumour with cytological uniformity <strong>and</strong> morphologicaldiversity [171]. The architecture comprises a variety <strong>of</strong>patterns, including ducts, streams, <strong>and</strong> micropapillary,cribriform <strong>and</strong> solid structures (Fig. 5.36). Diffuse infiltration<strong>of</strong> tumour cells with Indian filing <strong>and</strong> concentricgrowth around nerves is reminiscent <strong>of</strong> lobular carcinoma<strong>of</strong> <strong>the</strong> breast (Fig. 5.37).The cells each have single regular round, ovoid or fusiformbl<strong>and</strong> nuclei, sometimes with intra-nuclear vacuoles[187] <strong>and</strong> absent or small nucleoli. Variably presentare oncocytic, clear or mucous cells. Mitotic figuresare scanty, <strong>and</strong> never atypical. The stroma varies fromfibromyxoid to densely hyaline, but <strong>the</strong> chondroid matrix<strong>of</strong> a pleomorphic adenoma is not seen. Immunohistochemistryshows positivity with epi<strong>the</strong>lial markers(cytokeratins, EMA), S-100, bcl-2 <strong>and</strong> sometimes CEA,SMA <strong>and</strong> vimentin [26]; MIB1 proliferation is low –mean 2.4% (range 0.2–6.4) in one study [201]. PLGAbehaves as a low-grade malignancy; a literature reviewfound a recurrence rate <strong>of</strong> 21%, regional nodal metastasisin 6.5%, distant metastasis in 1.8%, <strong>and</strong> death due tocancer in 0.9% [116]. However, after 10 years late recurrences<strong>and</strong> metastases are perhaps more common thanthat [63], although in ano<strong>the</strong>r study with a long followup,recurrence was in large part due to incompleteness<strong>of</strong> excision – none <strong>of</strong> <strong>the</strong> 22 excised tumours recurredor caused death [159]. In a larger series <strong>of</strong> 164 PLGA,more than 95% <strong>of</strong> <strong>the</strong> patients had no evidence <strong>of</strong> diseaseafter a long-term follow-up [26]. The recommendedtreatment <strong>of</strong> PLGA is wide, but conservative surgicalexcision, postoperative radiation <strong>and</strong> chemo<strong>the</strong>rapyhave little place.The most important histopathological differential diagnosisis from <strong>the</strong> much more aggressive adenoid cysticcarcinoma. Although both are diffusely infiltratingcarcinomas that display morphological diversity, at a cytologicallevel <strong>the</strong> nuclei in AdCC are seen to be hyperchromatic,angulated, pleomorphic <strong>and</strong> densely packedwith more frequent mitotic figures, in contrast to <strong>the</strong>nuclei in PLGA, which are uniform with finely speckledchromatin. In addition, staining with S-100 protein isusually more diffuse <strong>and</strong> stronger in PLGA than AdCC[201, 225]. O<strong>the</strong>r markers such as c-kit (CD117) are <strong>of</strong>little use in practice, as staining can be seen in AdCC<strong>and</strong> most PLGAs [59]. Much more reliable marker is <strong>the</strong>MIB1 proliferation index, which is almost always significantlylower in PLGA [201, 225]. O<strong>the</strong>r differential diagnosesinclude pleomorphic adenoma, which in minorsalivary gl<strong>and</strong>s can be poorly circumscribed. The presence<strong>of</strong> chondroid matrix <strong>and</strong> any circumscription fa-


150 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5Fig. 5.38. Cribriform adenocarcinoma <strong>of</strong> <strong>the</strong> tongue (CAT). Avaguely nodular growth pattern is composed <strong>of</strong> solid nests withtubular structuresFig. 5.39. Epi<strong>the</strong>lial myoepi<strong>the</strong>lial carcinoma (EMCa): characteristicbiphasic appearance with an inner layer <strong>of</strong> ductal cells <strong>and</strong>outer layer <strong>of</strong> clear myoepi<strong>the</strong>lial cells. Basal membrane-like materialsurrounds <strong>the</strong> outer cellsvours PA, but it is sometimes not possible to distinguish<strong>the</strong>se tumours, particularly on a small biopsy. Papillarystructures form part <strong>of</strong> <strong>the</strong> spectrum <strong>of</strong> growth patternsseen in PLGA [206], but when extensive, <strong>the</strong>re isevidence that <strong>the</strong>se tumours are slightly but significantlymore aggressive [63, 64], although <strong>the</strong>y do not seemto affect long-term survival. Genuine high-grade malignancycan occur rarely, as ei<strong>the</strong>r a poorly differentiatedPLGA or as a salivary duct carcinoma [191].5.9.4.1 Cribriform Adenocarcinoma<strong>of</strong> <strong>the</strong> TongueICD-O:8525/3A newly described tumour [134] found so far only in <strong>the</strong>tongue, shares some histological features with PLGA,to which it is probably related. Cribriform adenocarcinoma<strong>of</strong> <strong>the</strong> tongue (CAT) usually arises in adults witha mean age <strong>of</strong> 50 years <strong>and</strong> equal sex incidence in <strong>the</strong>root <strong>of</strong> <strong>the</strong> tongue. Generally, at <strong>the</strong> time <strong>of</strong> diagnosis<strong>the</strong>re are metastases in <strong>the</strong> neck lymph nodes, ei<strong>the</strong>runilaterally or bilaterally, but distant spread has notbeen described.Microscopic examination shows lobules divided byfibrous septa, composed <strong>of</strong> areas with solid <strong>and</strong> microcysticgrowth patterns. In <strong>the</strong> solid areas, tumour nests<strong>of</strong>ten display a well-developed hyperchromatic outerlayer with a perpendicular arrangement <strong>of</strong> cells. Thislayer is frequently detached, forming papillae or glomeruloidstructures surrounded by apparent clefts. The microcysticgrowth pattern is composed <strong>of</strong> lobules <strong>of</strong> neoplasticcells with a cribriform <strong>and</strong>/or tubular architecture,<strong>the</strong> two patterns <strong>of</strong>ten intermingling. Typically, <strong>the</strong>tubules are approximately <strong>of</strong> <strong>the</strong> same size <strong>and</strong> consist <strong>of</strong>one cell layer. Cytologically, <strong>the</strong>re is one cell type; characteristically,<strong>the</strong> nuclei, which <strong>of</strong>ten overlap one ano<strong>the</strong>r,are pale <strong>and</strong> vesicular with a “ground glass” quality,thus resembling those <strong>of</strong> papillary thyroid carcinoma(Fig. 5.38).Each nucleus can contain up to three nucleoli <strong>of</strong> varyingconspicuousness. Immunohistochemically, a strongor patchy reaction is seen with cytokeratins <strong>and</strong> S-100protein. Actin, calponin <strong>and</strong> smooth muscle myosinheavy chain react with only a few areas. They are completelynegative for thyroglobulin.Patients treated with surgical excision <strong>and</strong> subsequentirradiation have a good chance <strong>of</strong> prolonged survivalwithout recurrence or fur<strong>the</strong>r metastatic spread[134].5.9.5 Epi<strong>the</strong>lial-Myoepi<strong>the</strong>lialCarcinomaICD-O:8562/3The mean age at diagnosis <strong>of</strong> epi<strong>the</strong>lial-myoepi<strong>the</strong>lialcarcinoma (EMCa) is 60 years (range 8–103), with asmall majority in females [62]. It occurs predominantlyin <strong>the</strong> parotid gl<strong>and</strong>, less <strong>of</strong>ten in <strong>the</strong> subm<strong>and</strong>ibulargl<strong>and</strong>, occasionally in minor salivary gl<strong>and</strong>s <strong>and</strong> rarelyin <strong>the</strong> bronchus [234]. The microscopic appearanceis characterised by small ductular lumina lined withtwo layers <strong>of</strong> cells (Fig. 5.39). The inner comprises cytokeratin-positiveepi<strong>the</strong>lial cells, <strong>and</strong> it is surroundedby an outer mantle <strong>of</strong> <strong>of</strong>ten clear myoepi<strong>the</strong>lial cells,which express SMA, smooth muscle myosin heavychain <strong>and</strong> calponin. S-100 protein also stains <strong>the</strong> outercells strongly, but is less specific <strong>and</strong> sometimes reactswith <strong>the</strong> inner layer [114] – CK 14 appears to be un-


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 151helpful. The outer cells are in turn surrounded by a rim<strong>of</strong> PAS-positive basement membrane material <strong>of</strong> variablethickness. This pattern is reproduced throughoutmost <strong>of</strong> <strong>the</strong> tumour, though each element may varyin prominence both between <strong>and</strong> within each lesion.Grossly, <strong>the</strong> tumours <strong>of</strong>ten appear to be well circumscribed,but microscopy usually reveals some invasion<strong>of</strong> surrounding structures. Cytological pleomorphismis infrequent, but mitotic figures may be numerous.The stroma is usually scanty, but on occasions it consists<strong>of</strong> plentiful hyaline basement membrane materialwith relatively inconspicuous bilayered ducts; <strong>the</strong> tumourcan <strong>the</strong>n be mistaken for a pleomorphic adenoma[186]. O<strong>the</strong>r differential diagnoses encompass a widerange <strong>of</strong> salivary neoplasms, mainly those composed <strong>of</strong>clear cells, both primary <strong>and</strong> metastatic. Many salivarytumours can be diagnosed purely on H&E morphology,but clear cell lesions are an exception, <strong>and</strong> most requireimmunohistochemistry <strong>and</strong> sometimes electronmicroscopy. EMCa can occasionally dedifferentiateas a high-grade adenocarcinoma [2] or a sarcomatoidspindle cell neoplasm <strong>of</strong> myoepi<strong>the</strong>lial type [186]. That<strong>the</strong>y originate from intercalated ducts is supported byan unusual case <strong>of</strong> a typical EMCa in a parotid gl<strong>and</strong>,which also contained multiple nodules <strong>of</strong> intercalatedduct hyperplasia (Figs. 5.40–5.42) [48].This appearance may explain why EMCa is not infrequentin hybrid tumours [31, 37]. The behaviour <strong>of</strong>EMCa is generally considered to be low grade, <strong>and</strong> ina literature review <strong>of</strong> 67 cases, recurrences were notedin 31%, cervical lymph node metastasis in 18%, distantmetastasis in 7% <strong>and</strong> death due to tumour in 7% [13]. Incontrast, <strong>the</strong> series <strong>of</strong> Fonseca <strong>and</strong> Soares [71] found that50% <strong>of</strong> neoplasms recurred <strong>and</strong> 40% <strong>of</strong> patients died <strong>of</strong>cancer. The only morphological feature found to correlatewith a poor prognosis was nuclear atypia in morethan 20% <strong>of</strong> tumour cells. In ano<strong>the</strong>r study DNA analysishas shown that aneuploidy is associated with an increasedchance <strong>of</strong> recurrence [34].5.9.6 Hyalinising Clear Cell CarcinomaICD-O:8310/3Monomorphic clear cell carcinomas are ei<strong>the</strong>r epi<strong>the</strong>lialor myoepi<strong>the</strong>lial (clear cell malignant myoepi<strong>the</strong>lioma).The former, now known as hyalinising clear cellcarcinoma was first described by Škorpil in Czech <strong>and</strong>German [205] <strong>and</strong> was rediscovered recently [138, 193],but was not included in <strong>the</strong> revised WHO classification[171]. It usually arises in <strong>the</strong> minor gl<strong>and</strong>s <strong>and</strong> is <strong>of</strong> lowgrademalignancy. Microscopically, it is characterised bygroups <strong>and</strong> trabeculae <strong>of</strong> polygonal glycogen-rich cellsseparated by dense collagen b<strong>and</strong>s. At times, particularlyin <strong>the</strong> deeper parts <strong>of</strong> <strong>the</strong> tumours, <strong>the</strong> cells maylose <strong>the</strong>ir clarity when <strong>the</strong>ir cytoplasm appears weaklyFigs. 5.40–5.42. Intercalated duct hyperplasia <strong>of</strong> <strong>the</strong> parotidgl<strong>and</strong>. Top: Hyperplastic foci are composed <strong>of</strong> an inner layer <strong>of</strong>epi<strong>the</strong>lial cells surrounded by myoepi<strong>the</strong>lial cells with ample <strong>and</strong>clear cytoplasm. The former stain for CAM5.2 (lower) <strong>and</strong> <strong>the</strong> latterfor smooth muscle actin (middle)eosinophilic. Immunohistochemistry reveals positivitywith epi<strong>the</strong>lial markers (e.g. cytokeratin), but myoepi<strong>the</strong>lialmarkers (e.g. S-100 protein <strong>and</strong> SMA) are negative[138].5.9.7 Basal Cell AdenocarcinomaThis tumour has <strong>the</strong> architecture <strong>and</strong> cytology <strong>of</strong> basalcell adenoma, but displays infiltration. Most cases arise


152 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5Fig. 5.43. Basal cell adenocarcinoma. In spite <strong>of</strong> <strong>the</strong> lack <strong>of</strong> significantcellular atypia, <strong>the</strong> infiltrative pattern is diagnostic <strong>of</strong> malignancy.Courtesy <strong>of</strong> Pr<strong>of</strong>. J.W. Eveson, Bristol, UKamount <strong>of</strong> basement membrane material varies, butcan be marked, especially in <strong>the</strong> membranous variant.Occasional cases show cytological pleomorphism, butgenerally this is absent, <strong>and</strong> mitotic figures are usuallysparse. The most reliable indicator <strong>of</strong> malignancy is infiltration<strong>of</strong> <strong>the</strong> surrounding gl<strong>and</strong>, <strong>and</strong> less frequently<strong>of</strong> blood vessels <strong>and</strong> nerves (Fig. 5.43) [16]. In addition,<strong>the</strong> Ki-67 proliferation index is usually higher in basalcell adenocarcinoma than its benign counterpart (>5%vs.


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 153Fig. 5.45. Myoepi<strong>the</strong>lial carcinoma, solid growth pattern withcentral necrosis in one <strong>of</strong> <strong>the</strong> nodules. This finding may mimicsalivary duct carcinoma with comedo-like necrosisFig. 5.47. Myoepi<strong>the</strong>lial carcinoma: focal squamous metaplasiawith keratin pearl formationFig. 5.46. Myoepi<strong>the</strong>lial carcinoma. The spindle cell componentshows nuclear pleomorphism resembling a s<strong>of</strong>t tissue sarcoma. Ahelpful diagnostic pointer is that o<strong>the</strong>r types <strong>of</strong> myoepi<strong>the</strong>lial cellare usually identified elsewhereinates, but <strong>the</strong>re is usually a minor component <strong>of</strong> o<strong>the</strong>rcell types. No true gl<strong>and</strong>s or lumina are seen in pure,malignant myoepi<strong>the</strong>liomas, but as with <strong>the</strong>ir benigncounterparts, occasional small ducts in a neoplasm witho<strong>the</strong>rwise typical features should not preclude <strong>the</strong> diagnosis[183]. The nuclei can vary from relatively uniform,small with finely distributed chromatin, lacking obviousnucleoli, to markedly enlarged <strong>and</strong> pleomorphic, showingchromatin clumping <strong>and</strong> large nucleoli. Mitotic figuresmay be plentiful (range 3 to 51 per 10 high powerfields) <strong>and</strong> include atypical forms [168]. Multinucleate[33] <strong>and</strong> bizarre tumour giant cells may occasionally bepresent. The tumour-related matrix is generally prominent<strong>and</strong> is hyalinised or myxoid.Special stains in tumours without any ductal differentiationshow no mucicarmine-positive mucus,but plentiful glycogen is found in clear cells <strong>and</strong> <strong>the</strong>myxoid matrix is positive with Alcian Blue. Metaplasticchanges are frequent <strong>and</strong> include areas showingsquamous differentiation, <strong>of</strong>ten with keratinisation(Fig. 5.47). Perineural invasion is seen in 44% <strong>and</strong> vascularinvasion in 16%. In one series, 40% <strong>of</strong> tumourswere categorised as high grade <strong>and</strong> 60% as low grade[168]. All tumours show some positivity for S-100 protein,vimentin <strong>and</strong> broad-spectrum cytokeratin (e.g.AE1-AE3 or MNF116). O<strong>the</strong>r cytokeratin antisera(CAM 5.2 <strong>and</strong> LP34) show some reactivity in most tumours,<strong>and</strong> about half display some expression <strong>of</strong> cytokeratin[13]. Of <strong>the</strong> more specific myoepi<strong>the</strong>lial markers,approximately 75% <strong>of</strong> tumours including thosecomposed <strong>of</strong> plasmacytoid cells, express calponin <strong>and</strong>about 50% are positive with αSMA; p63 was positivein 60% [166]. Amongst o<strong>the</strong>r markers, glial fibrillaryacidic protein (GFAP) is positive in 31% <strong>and</strong> epi<strong>the</strong>lialmembrane antigen (EMA) in 20%, in addition tohighlighting any true small ducts, but carcino-embryonicantigen (CEA) is usually negative. CD117 (c-kit)was positive in <strong>the</strong> few cases studied [112]. The meanMIB1 (Ki-67) index in one series was 35% (range 15–65), with any count above 10% said to be diagnostic <strong>of</strong>malignancy in a myoepi<strong>the</strong>lial neoplasm [151].Electron microscopy shows that some tumour cellscontain small desmosomes, but actin filaments are few[168]. It has been shown that malignant myoepi<strong>the</strong>liomassecrete matrix-degrading proteinases, as well asproteinase inhibitors [215], <strong>and</strong> this appears to be associatedwith demonstrated inhibition <strong>of</strong> angiogenesis.These features indicate an anti-invasive effect, <strong>and</strong> althoughas yet poorly understood, this is likely to have


154 S. Di Palma · R.H.W. Simpson · A. Skalova · I. LeivoTable 5.3. Classification <strong>of</strong> clear cell tumours <strong>and</strong> tumour-like conditions <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s [174]5BenignMalignant, primary(carcinomas not usually characterised byclear cells, but with rare clear cell variants)Malignant, primary(carcinomas usually characterised by clearcells)Malignant, metastaticPleomorphic adenomaMyoepi<strong>the</strong>liomaSebaceous adenomaOncocytomaMultifocal nodular oncocytic hyperplasia (MNOH)Mucoepidermoid carcinomaAcinic cell carcinomaEpi<strong>the</strong>lial-myoepi<strong>the</strong>lial carcinomaHyalinising clear cell carcinomaClear cell malignant myoepi<strong>the</strong>lioma (myoepi<strong>the</strong>lial carcinoma)Sebaceous carcinomaCarcinomas: especially kidney, thyroid. Also melanomaan effect on <strong>the</strong> biological aggressiveness <strong>of</strong> any particulartumour.The variable appearance <strong>of</strong> malignant myoepi<strong>the</strong>lioma,leads to a wide differential diagnosis, includingo<strong>the</strong>r salivary carcinomas. Nodules with central necrosismimic <strong>the</strong> comedocarcinoma structures in salivaryduct carcinoma, but <strong>the</strong>re is usually more stromal materialin <strong>the</strong> myoepi<strong>the</strong>lial neoplasm <strong>and</strong> in addition, S-100 <strong>and</strong>/or myoepi<strong>the</strong>lial markers are usually positive.The clear cell variant resembles many o<strong>the</strong>r salivaryneoplasms composed <strong>of</strong> clear cells, benign <strong>and</strong> malignant,primary <strong>and</strong> metastatic (Table 5.3) [132, 174] (seeSect. 5.9.5).The prognosis <strong>of</strong> malignant myoepi<strong>the</strong>lioma is variable,but approximately one-third <strong>of</strong> patients die <strong>of</strong> disease,ano<strong>the</strong>r third have residual tumour <strong>and</strong> <strong>the</strong> remainingthird are disease-free [62, 168]. When metastasesoccur, <strong>the</strong>y can be found in neck lymph nodes<strong>and</strong> at distant sites, including lungs, kidney, brain <strong>and</strong>bones. Malignant myoepi<strong>the</strong>liomas arising in ordinarypleomorphic adenomas behave in <strong>the</strong> same way as thosethat arise de novo [168], but it has been suggested thatneoplasms developing in multiply recurrent pleomorphicadenomas may pursue a prolonged course [52]. Afur<strong>the</strong>r suggestion is that malignant myoepi<strong>the</strong>liomascomposed mainly <strong>of</strong> plasmacytoid cells may be more aggressive[53, 218]. However, Savera <strong>and</strong> Sloman in <strong>the</strong>irseries <strong>of</strong> 25 cases found only a weak statistical correlationfor outcome with cytological atypia (high grade),but o<strong>the</strong>r parameters (tumour size, site, cell type, mitoticrate, presence <strong>of</strong> a benign tumour, necrosis, perineural<strong>and</strong> vascular invasion) showed no relationship [168].In practice with any particular case, <strong>the</strong> various histologicalfeatures should be listed, <strong>and</strong> an attempt made todescribe <strong>the</strong> tumour as low- or high-grade, but adding arider that histological grade is as yet a far from provenguide to clinical behaviour. Treatment is surgical, <strong>and</strong>no role for radio- <strong>and</strong> chemo<strong>the</strong>rapy has yet been established.5.9.9 Salivary Duct CarcinomaICD-O:8500/3Salivary duct carcinoma (SDC) is probably not as uncommonas previously thought [87, 93]. Most patientsare over 50 years old <strong>and</strong> <strong>the</strong>re is at least a 3:1 male predominance.It arises mainly in <strong>the</strong> parotid, less <strong>of</strong>ten in<strong>the</strong> subm<strong>and</strong>ibular gl<strong>and</strong> <strong>and</strong> only occasionally in <strong>the</strong>minor gl<strong>and</strong>s <strong>of</strong> <strong>the</strong> palate [47], buccal mucosa [162],maxilla [122] <strong>and</strong> larynx [69]. It can develop de novoor in a pre-existing pleomorphic adenoma [86, 87] orpolymorphous low-grade adenocarcinoma [191]. Themicroscopic appearance <strong>of</strong> SDC bears a striking resemblanceto ductal carcinoma <strong>of</strong> <strong>the</strong> breast, both in situ<strong>and</strong> invasive, where all <strong>of</strong> <strong>the</strong> features <strong>of</strong> <strong>the</strong> mammaryequivalent can be reproduced (Fig. 5.48). Perineural <strong>and</strong>lymphovascular invasion are seen in many cases. Nuclearpleomorphism is usual, <strong>and</strong> is also apparent on FNAcytology [192]. Mitotic figures are <strong>of</strong>ten numerous, alsoreflected in a high Ki-67 proliferation index [93].Although most cases <strong>of</strong> SDC can be diagnosed withHE alone, special stains can help in a few instances. Immunohistochemistryshows expression <strong>of</strong> epi<strong>the</strong>lial markerssuch as cytokeratins (including subtype 7, but not 20),EMA <strong>and</strong> GCDFP-15. S-100 protein is usually negative, asare myoepi<strong>the</strong>lial markers, although <strong>the</strong>y may highlightsubtle in situ lesions [5]. Despite <strong>the</strong> morphological similarityto breast carcinoma, staining for oestrogen <strong>and</strong> progesteronereceptors is almost always negative, in contrastto that for <strong>and</strong>rogen receptors (AR), where >90% <strong>of</strong> SDCsreact, even in women. AR positivity appears to be specificto SDC (including when it arises in a pleomorphic adenoma),<strong>and</strong> is not seen in for example, mucoepidermoid


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 155Fig. 5.48. Salivary duct carcinoma: invasive irregular ducts <strong>and</strong>cribriform structures strongly resemble ductal carcinoma <strong>of</strong> <strong>the</strong>breastFig. 5.49. Salivary duct carcinoma, mucin-rich variant. This iscomposed <strong>of</strong> a mixture <strong>of</strong> usual-type salivary duct carcinoma <strong>and</strong>lakes <strong>of</strong> mucinous adenocarcinomacarcinoma [142]. Some studies have shown SDCs to expressprostate specific antigen (PSA) or acid phosphatase[110, 120], but ano<strong>the</strong>r failed to confirm this [185] <strong>and</strong>similarly, only 1 out <strong>of</strong> 40 cases in a Mayo Clinic serieswas PSA-positive [115]. More recently cases <strong>of</strong> SDC showingpositive staining for HER-2/neu (c-erbB-2) protein onimmunohistochemistry have been published [202], <strong>and</strong><strong>the</strong> gene amplification has been demonstrated with FISHanalysis [204].Several rare morphological variants <strong>of</strong> SDC havebeen described, including cribriform [23], micropapillary[147], sarcomatoid [96], mucin-rich [190] <strong>and</strong> oncocytic(Figs. 5.49, 5.50) [184]. So-called low grade salivaryduct carcinoma [46] is probably a separate entity(see below).The differential diagnoses <strong>of</strong> SDC are high-grademucoepidermoid carcinoma, oncocytic carcinoma <strong>and</strong>some metastases. The diagnosis <strong>of</strong> mucoepidermoidcarcinoma requires <strong>the</strong> presence <strong>of</strong> squamous-like cells,mucus-producing cells <strong>and</strong> cells <strong>of</strong> intermediate type,<strong>and</strong> <strong>the</strong>re is no expression <strong>of</strong> <strong>and</strong>rogen receptors. Manysalivary oncocytic carcinomas probably demonstrateo<strong>the</strong>r types <strong>of</strong> malignancy (including SDC) with plentifuloncocytic cells, but a true oncocytic carcinoma lacksany features <strong>of</strong> SDC <strong>and</strong> is AR-negative. At present, nei<strong>the</strong>r<strong>of</strong> <strong>the</strong>se two differential diagnoses is clinically criticalsince <strong>the</strong> prognosis is similar. However, it is importantto identify metastatic carcinoma, particularly from<strong>the</strong> prostate or breast. In most cases, metastases will beobvious from clinical investigation <strong>and</strong> imaging studies,but <strong>the</strong> usual immunopr<strong>of</strong>ile <strong>of</strong> <strong>the</strong>se tumours is different:prostatic carcinomas tend to be AR+, ER−, PSA+,CK 7+; breast carcinoma tend to be usually AR−, <strong>of</strong>tenER +, PSA−, CK 7+; SDC is AR+, ER−, usually PSA−, <strong>and</strong>CK 7+.Fig. 5.50. Salivary duct carcinoma with oncocytic differentiation.The cells have ample granular cytoplasm with vesicular nuclei<strong>and</strong> prominent nucleoli. A clear distinction between oncocyticsalivary duct carcinoma <strong>and</strong> true oncocytic carcinoma may notbe possible, as <strong>the</strong>y may not be separate entitiesThe prognosis for SDC is poor, <strong>and</strong> most series haveshown that more than 70% <strong>of</strong> patients die <strong>of</strong> disease,usually within 3 years. Never<strong>the</strong>less, Grenko et al. [86]alluded to a minority (about 25–30%) who do well, but<strong>the</strong>ir study was unable to identify any particular features<strong>of</strong> this group. Amongst possible prognostic indicators,tumour size is probably important, with lesions


156 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5with radical neck dissection followed by radio<strong>the</strong>rapy to<strong>the</strong> tumour bed <strong>and</strong> chemo<strong>the</strong>rapy [36, 145]. The recentidentification <strong>of</strong> <strong>and</strong>rogen receptors expression in SDCraises <strong>the</strong> question as to whe<strong>the</strong>r anti-<strong>and</strong>rogen <strong>the</strong>rapy(e.g. flutamide or goserelin) might have merit [99,142, 153]. The significance <strong>of</strong> staining for HER-2/neu (cerbB-2)protein is at present uncertain [202, 204], ei<strong>the</strong>rprognostically or for planning <strong>the</strong>rapy with herceptin.The entity “low grade salivary duct carcinoma (lowgrade cribriform cystadenocarcinoma)” has a predominantlyintraductal growth pattern with low-grade cytologicalfeatures [46], <strong>and</strong> its relationship to usual-typeSDC is as yet uncertain. The tumour is S-100-positive<strong>and</strong> <strong>the</strong> only case studied for AR was negative [19]. Theprognosis is good, <strong>and</strong> although only a few cases havebeen described, none <strong>of</strong> <strong>the</strong> patients has died <strong>of</strong> disease.5.9.10 Oncocytic CarcinomaICD-O:8290/3Several carcinoma types have variants composed <strong>of</strong>oncocytic cells, but only a few dozen cases <strong>of</strong> pure oncocyticcarcinoma have been reported. The averageage is 63 years (range 29–91), <strong>and</strong> most have occurredin <strong>the</strong> parotid [62]; some have arisen in Warthin’s tumours[222]. The diagnosis <strong>of</strong> a pure oncocytic carcinomarequires <strong>the</strong> identification <strong>of</strong> malignancy,oncocytic differentiation <strong>and</strong> lack <strong>of</strong> features <strong>of</strong> anyo<strong>the</strong>r tumour type. It is likely that a pure oncocyticcarcinoma is an aggressive tumour, as over half <strong>of</strong> <strong>the</strong>patients reported ei<strong>the</strong>r died <strong>of</strong> disease or suffered recurrences[169].5.9.11 Malignancy in PleomorphicAdenoma MalignantMixed TumourThe histological diagnosis <strong>of</strong> pleomorphic adenoma(PA; benign mixed tumour) is not always straightforward,as benign lesions may display atypical histologicfeatures such as capsular infiltration, hypercellularity,cellular atypia, necrosis <strong>and</strong> vascular invasion [3, 7],which cause suspected malignancy. In addition, somePAs contain genuine cytologically malignant cells, butbehave in a benign fashion [21, 56]. A fur<strong>the</strong>r paradoxis <strong>the</strong> rare occurrence <strong>of</strong> histologically benign-lookingPAs that metastasise [232]. Thus, <strong>the</strong> concept <strong>of</strong> malignancyin PA is much more complex than appears at firstsight. This is reflected by <strong>the</strong> variable incidence for <strong>the</strong>reported frequency <strong>of</strong> malignancy in PA. In several largeseries [83, 125, 149, 209, 223] <strong>the</strong> average was 3.6% <strong>of</strong>all salivary gl<strong>and</strong> tumours <strong>and</strong> 11.7% <strong>of</strong> malignancies;overall, malignancy develops in 6.2% <strong>of</strong> all PAs (rangein different series 1.9 to 23.3%). The incidence <strong>of</strong> malignantchange increases with <strong>the</strong> length <strong>of</strong> history <strong>of</strong><strong>the</strong> PA, from 1.5% at 5 years to 10% after 15 years. Theconcept <strong>of</strong> malignant mixed tumour (MMT) as a malignanttumour that contains remnants <strong>of</strong> benign mixedtumour was developed by LiVolsi <strong>and</strong> Perzin in 1977[125]. Spiro et al. [209] agreed with this, but suggesteda possible de novo origin in cases lacking a clinical historyor histological evidence <strong>of</strong> a pre-existing salivarygl<strong>and</strong> tumour.The revised WHO classification <strong>of</strong> salivary gl<strong>and</strong> tumours[171] discussed <strong>the</strong> topic <strong>of</strong> MMT with <strong>the</strong> titleCarcinoma in Pleomorphic Adenoma <strong>and</strong> <strong>the</strong> sub-titleMalignant Mixed Tumour. Three entities are recognised:carcinoma in pleomorphic adenoma, carcinosarcoma(true malignant mixed tumour) <strong>and</strong> metastasising pleomorphicadenoma [171]. There is no uniform agreementon this classification as metastasising PA does not containhistological features <strong>of</strong> malignancy <strong>and</strong> <strong>the</strong>refore it isanomalous to include it as a form <strong>of</strong> malignancy in PA. Inaddition, not all carcinosarcomas arise from a PA. Finally,<strong>the</strong> possibility <strong>of</strong> myoepi<strong>the</strong>lial malignancy arising in aPA is not included [49].5.9.11.1 CarcinomaEx Pleomorphic AdenomaICD-O:8941/3Two subtypes should be recognised: invasive <strong>and</strong> noninvasivecarcinoma.5.9.11.1.1 InvasiveCarcinoma Ex PAThis is <strong>the</strong> commoner form, in which <strong>the</strong> malignancyinvolves only <strong>the</strong> epi<strong>the</strong>lial component. It occurs mainlyin men over 60 years old. Most cases (81.7%) involve <strong>the</strong>parotid gl<strong>and</strong>, with <strong>the</strong> subm<strong>and</strong>ibular in 18% <strong>and</strong> <strong>the</strong>sublingual in 0.3%; <strong>the</strong> minor salivary gl<strong>and</strong>s, particularlyin <strong>the</strong> palate, can also be affected [229]. The typicalpresentation is a long history <strong>of</strong> a salivary gl<strong>and</strong> nodulethat suddenly increases in size.The demonstration <strong>of</strong> both a carcinoma <strong>and</strong> a PA isnecessary for <strong>the</strong> diagnosis.A history <strong>of</strong> a long-st<strong>and</strong>ing parotid tumour is notsufficient evidence for a pre-existing PA, whilst a previouslyexcised PA at <strong>the</strong> site <strong>of</strong> a carcinoma is acceptable[62].Grossly, carcinoma ex PA is <strong>of</strong>ten larger than a benignPA. Histological recognition <strong>of</strong> a pre-existing PAmay be difficult, as it could be obscured by <strong>the</strong> carcinoma,or may only show degenerate changes such as


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 157Fig. 5.51. Non-invasive carcinoma in a pleomorphic adenoma.Ducts contain cells with atypical nuclei. Focal necrosis <strong>and</strong> calcificationis also presentFig. 5.52. True malignant mixed tumour/carcinosarcoma. Theepi<strong>the</strong>lial component is a poorly differentiated carcinoma withsome features suggesting salivary duct carcinoma. The sarcomatouscomponent is a high-grade spindle cell sarcoma, in this case,without specific differentiationscarring, dystrophic calcification, necrosis <strong>and</strong> haemorrhagewith occasional transitional changes made up<strong>of</strong> cells showing features intermediate between frankmalignancy <strong>and</strong> PA [49, 124]. Although <strong>the</strong> proportion<strong>of</strong> <strong>the</strong> malignant component varies from minutefoci to almost <strong>the</strong> whole lesion, recognition <strong>of</strong> franklyinvasive carcinoma ex PA is usually simple. Capsular,perineural <strong>and</strong> vascular invasion are easily identified,as well as extension into neighbouring tissues [49,62, 124].Recent studies show adenocarcinoma not o<strong>the</strong>rwisespecified <strong>and</strong> salivary duct carcinoma to be <strong>the</strong>most frequent histological types [49, 87], <strong>and</strong> <strong>the</strong>re isimmunohistochemical <strong>and</strong> ultrastructural evidencethat many carcinomas previously described as “undifferentiated”are in fact myoepi<strong>the</strong>lial – indeed manymyoepi<strong>the</strong>lial carcinomas can be shown to have arisenin a pre-existing PA [52, 53, 151, 168]. It is not uncommonto find o<strong>the</strong>r concurrent differentiation,e.g. squamous, mucoepidermoid, polymorphous lowgradeadenocarcinoma [125, 149, 209]. In determining<strong>the</strong> prognosis, <strong>the</strong> extent <strong>of</strong> invasion is more importantthan <strong>the</strong> histological type: Tortoledo et al. [223]found that none <strong>of</strong> <strong>the</strong> patients whose tumour penetrated8 mm died <strong>of</strong> disease.A more recent study found that none <strong>of</strong> <strong>the</strong> tumoursthat invaded


158 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5<strong>the</strong> history will usually be that <strong>of</strong> rapid growth in a longst<strong>and</strong>ingsalivary nodule.Microscopy shows a biphasic tumour composed <strong>of</strong>epi<strong>the</strong>lial <strong>and</strong> mesenchymal elements. The former isgenerally a poorly differentiated (adeno)carcinoma,but salivary duct carcinoma is increasingly reported(Fig. 5.52) [49, 50]. The o<strong>the</strong>r component is usuallya chondrosarcoma, but osteogenic sarcoma, fibrosarcoma,malignant fibrous histiocytoma, pleomorphicrhabdomyosarcoma <strong>and</strong> osteoclast-type giant cellneoplasms [214, 224] have also been described.Epi<strong>the</strong>lial markers are usually detected in <strong>the</strong> epi<strong>the</strong>lialcomponent, which may or may not also be expressedin <strong>the</strong> sarcomatous component. Positive stainingfor epi<strong>the</strong>lial markers has been used as pro<strong>of</strong> <strong>of</strong> <strong>the</strong>fact that CS are carcinomas showing divergent differentiation<strong>and</strong> as an indication <strong>of</strong> <strong>the</strong>ir monoclonal origin.However, keratin staining can be negative castingdoubt onto <strong>the</strong> monoclonal-carcinomatous nature <strong>of</strong><strong>the</strong> whole tumour. Molecular studies have been helpfulin clarifying this issue. In analogous neoplasmsin o<strong>the</strong>r organs such as breast, uterus <strong>and</strong> in salivarygl<strong>and</strong>s, molecular studies have demonstrated that carcinomatous<strong>and</strong> sarcomatous components have similargenetic pr<strong>of</strong>iles. Moreover, in a subset <strong>of</strong> CS withosteoclastic-type giant cells, Tse et al. [224] found mutation<strong>of</strong> <strong>the</strong> same allele on chromosome 17p13, whichis a known mutation <strong>of</strong> salivary duct carcinoma. Thisindicates that carcinosarcoma are in fact carcinomas<strong>of</strong> high-grade malignancy <strong>and</strong> should be treated assuch. HER-2 overexpression on immunohistochemistryis also seen in <strong>the</strong> salivary duct component <strong>of</strong> CS.The meaning <strong>of</strong> this information has still not beenclarified [57].5.9.11.3 MetastasisingPleomorphic AdenomaICD-O:8940/0This tumour is histologically indistinguishable frombenign PA, yet it metastasises widely to sites includinglymph nodes, bone, lung <strong>and</strong> kidney, <strong>and</strong> can kill <strong>the</strong>patient [232]. Whereas <strong>the</strong> WHO revised classificationlists metastasising pleomorphic adenoma (MPA)as one entity in <strong>the</strong> MMT category <strong>of</strong> [171], it differsbecause it remains histologically “benign” in <strong>the</strong> primarysite, local recurrences, <strong>and</strong> metastatic deposits[56, 232].It is a rare tumour with fewer than 100 reported casesso far. Despite this, MPA has a clear-cut clinicopathologicalpr<strong>of</strong>ile: <strong>the</strong> reported cases shared several similarities,such as long time intervals (up to 50 years) between<strong>the</strong> primary tumour <strong>and</strong> metastases, <strong>and</strong> simultaneous,usually multiple, local recurrences <strong>and</strong> distantmetastases [232]. Although <strong>the</strong> morphology <strong>of</strong> both isalmost identical, <strong>the</strong> recurrences seem to play an importantrole in <strong>the</strong> genesis <strong>of</strong> systemic spread. This suggeststhat surgical manipulation may favour vascularimplantation or invasion eventually leading to metastases,but in many cases <strong>of</strong> MPA it was not possible histologicallyto demonstrate actual vascular permeation[56, 232].5.9.12 Sebaceous CarcinomaICD-O:8410/3Although sebaceous gl<strong>and</strong>s are common in <strong>the</strong> oral mucosa(Fordyce granules), sebaceous neoplasms <strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong>s are rare. Most sebaceous carcinomas havearisen in <strong>the</strong> parotid [62], possibly from pluripotent ductcells [219]. The sex incidence is equal, <strong>and</strong> <strong>the</strong> meanage is 69 years (range 17–93). Macroscopically, <strong>the</strong>y arepartly encapsulated <strong>and</strong> vary in size from 6 to 85 mmacross <strong>the</strong> greatest diameter. Microscopy shows invasiveisl<strong>and</strong>s, duct-like structures <strong>and</strong> sheets <strong>of</strong> tumourcells, which may be sebaceous, squamous or basaloid;intracellular mucin may be found [12]. Sebaceous cellsare present in varying numbers, <strong>and</strong> typically comprisefoamy cytoplasm <strong>and</strong> a single vesicular nucleus witha prominent nucleolus. Areas <strong>of</strong> necrosis are frequent[85]. The tumour cells react with cytokeratin <strong>and</strong> EMA,but not with S-100 protein or actin [219]. The behaviouris intermediate to high-grade, <strong>and</strong> recurrences, metastases<strong>and</strong> death due to disease have all been reported.Three cases <strong>of</strong> sebaceous lymphadenocarcinoma havebeen described, representing malignant transformation<strong>of</strong> sebaceous lymphadenoma. One <strong>of</strong> <strong>the</strong> patients diedbecause <strong>of</strong> <strong>the</strong> tumour [62].5.9.13 Lymphoepi<strong>the</strong>lial CarcinomaICD-O:8082/3The WHO revised classification includes this tumourunder undifferentiated carcinomas [171], but it is a genuineclinicopathological entity <strong>and</strong> can be consideredseparately.Lymphoepi<strong>the</strong>lial carcinoma is extremely rare exceptin Eskimos (Inuit) <strong>and</strong> in Sou<strong>the</strong>rn China. Themedian age is 40 years (range 10–86), <strong>and</strong> it is slightlycommoner in females [62]; familial clusters have beenidentified amongst patients from Greenl<strong>and</strong> [1]. Theparotid is involved in 80% <strong>of</strong> cases, with <strong>the</strong> rest occurringin <strong>the</strong> subm<strong>and</strong>ibular gl<strong>and</strong>s. Forty per cent <strong>of</strong> patientshave lymph node metastases at <strong>the</strong> time <strong>of</strong> presentation.A few examples have been described in associationwith lymphoepi<strong>the</strong>lial sialadenitis [121], but amuch more important association is with Epstein-Barr


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 159Table 5.4. Metastases to <strong>the</strong> parotid gl<strong>and</strong>, adapted from Gnepp [82]Location <strong>of</strong> primaryNumber <strong>of</strong> tumoursSkin <strong>of</strong> head <strong>and</strong> neck 422 (53.8%)Upper aero-digestive tract (mouth, nose, sinuses, pharynx) 63Eye (conjunctiva, lacrimal gl<strong>and</strong>) 6Thyroid 5<strong>Head</strong>, not o<strong>the</strong>rwise specified 4Central nervous system 4Subm<strong>and</strong>ibular salivary gl<strong>and</strong> 1Lung 28Kidney 23Breast 19Colorectal 7Prostate 4Skin, distant 3Stomach 2Uterus 1Pancreas 1Total, distant sites 88 (11.2%)Skin, not o<strong>the</strong>rwise specified 108Unknown primary site 84virus, <strong>and</strong> viral genomes can be detected in <strong>the</strong> malignantcells [89].There is a marked histological similarity to undifferentiatednasopharyngeal carcinoma, which has alsobeen linked to Epstein-Barr virus. Microscopic examinationshows syncytial groups <strong>of</strong> large epi<strong>the</strong>lialcells with vesicular nuclei <strong>and</strong> prominent nucleoli,intimately mixed with lymphocytes <strong>and</strong> plasma cells,sometimes with germinal centre formation. Mitoticfigures are <strong>of</strong>ten numerous. At times, <strong>the</strong> epi<strong>the</strong>liumis difficult to identify, but it can be highlighted by cytokeratinmarkers. The most important differential diagnosisis a metastasis from a nasopharyngeal primary,which can present as a parotid mass [231], or possiblyvery poorly differentiated squamous carcinoma <strong>of</strong>usual type originating in <strong>the</strong> skin or upper aerodigestivetract. The outcome is surprisingly good for suchan aggressive-looking carcinoma, <strong>and</strong> <strong>the</strong> 5-year survivalrate is 60%.5.9.14 Small Cell CarcinomaICD-O:8041/3Small cell carcinoma (SCC) is unlikely to be a singleentity, as electron microscopy reveals that some neoplasmsshow neuroendocrine differentiation, whilsto<strong>the</strong>rs have squamous <strong>and</strong> ductal features not apparenthistologically [15, 118], <strong>and</strong> occasionally both patternsare evident in <strong>the</strong> same tumours. Some neoplasmscalled small cell carcinoma may in fact be primaryprimitive neuroectodermal tumours [44]. They are seenmore <strong>of</strong>ten in men, <strong>and</strong> <strong>the</strong> mean age is 56 years (range5–86) [62]. The microscopic appearance may be similarto small cell carcinoma <strong>of</strong> <strong>the</strong> lung or Merkel cell carcinoma<strong>of</strong> <strong>the</strong> skin. Both comprise solid sheets, nests <strong>and</strong>cords <strong>of</strong> closely packed cells; <strong>the</strong> difference is in <strong>the</strong> cellsize – small <strong>and</strong> dark cells in <strong>the</strong> former, slightly larger<strong>and</strong> with pale chromatin in <strong>the</strong> latter.Immunohistochemistry shows positive staining forchromogranin, synaptophysin, neuron-specific enolase<strong>and</strong> CAM5.2, <strong>of</strong>ten with paranuclear dots in bothtypes. However, immunohistochemistry for cytokeratin20 seems to identify two subtypes <strong>of</strong> small cell carcinoma:CK 20− lung cell type <strong>and</strong> CK 20+ Merkel celltype carcinoma. A recent study by Nagao et al. [148]showed that CK 20+ small cell carcinomas <strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong>s have a better prognosis than CK 20− cases.This suggests that staining for CK 20 should beperformed in SCC as <strong>the</strong> results may have prognosticvalue [148]. The differential diagnosis includes metastasisfrom small cell carcinomas <strong>of</strong> <strong>the</strong> lung <strong>and</strong> thismust be excluded before a primary small cell carcinomacan be said to be <strong>of</strong> salivary origin. Lymphomas<strong>and</strong> primary primitive neuroectodermal tumours <strong>of</strong><strong>the</strong> salivary gl<strong>and</strong>s [44, 105] may be somewhat similarmorphologically, <strong>and</strong> can be excluded immunohistochemically.


160 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5High-grade transformation is a rare but well-establishedevent in several primary low-grade salivary carcinomas,<strong>and</strong> usually heralds a more aggressive clinicalcourse. Examples have been described in acinic cell[51, 95, 154, 213], adenoid cystic [32, 140], epi<strong>the</strong>lialmyoepi<strong>the</strong>lial[2, 73, 186], mucoepidermoid [146] <strong>and</strong>polymorphous low-grade adenocarcinoma [160, 191],as well as malignant myoepi<strong>the</strong>lioma [156]. In eachcase, <strong>the</strong> diagnoses <strong>of</strong> high-grade change were basedon histopathological criteria, especially increased mitotic<strong>and</strong> proliferation rates. Most <strong>of</strong> <strong>the</strong> transformedcomponents were poorly differentiated adenocarcinomas,but some <strong>of</strong> those in adenoid cystic <strong>and</strong> epi<strong>the</strong>lial-myoepi<strong>the</strong>lialcarcinomas showed myoepi<strong>the</strong>lialfeatures. The processes underlying dedifferentiation <strong>of</strong>salivary neoplasms remain to be established, but previousradio<strong>the</strong>rapy may have been important in some <strong>of</strong><strong>the</strong> AdCCs [32] <strong>and</strong> PLGAs [160]. In general, no definitefactors in <strong>the</strong> progression <strong>of</strong> low-grade to highgradecarcinomas have been identified at a molecularlevel [208]; for example, <strong>the</strong>re is conflicting evidenceregarding p53 mutations, which might have been involvedin a single case <strong>of</strong> transformed AdCC [32], butwas not a factor in dedifferentiated acinic cell carcinomas[51, 95].5.9.16 MetastaticMalignanciesFigs. 5.53, 5.54. Endodermal sinus tumour <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>.Positive staining for placental alkaline phosphatase (PLAP) is essentialto confirm <strong>the</strong> diagnosis. Courtesy <strong>of</strong> Dr. Isabela Wernicke[211]5.9.15 Higher Grade Changein CarcinomasMetastases to <strong>the</strong> major gl<strong>and</strong>s <strong>and</strong> <strong>the</strong> intraparotidlymph nodes constitute approximately 10% <strong>of</strong> all salivarycarcinomas [82]; <strong>the</strong> exact figure varies fromstudy to study depending on local factors such as differentincidences <strong>of</strong> particular cancers. For example,Bergensen et al. [18] in Australia reported that metastasesconstituted 72% <strong>of</strong> all malignancies, resultingfrom <strong>the</strong> high incidence <strong>of</strong> skin cancer. In an AFIP series<strong>and</strong> literature review in 1991 <strong>of</strong> 785 parotid metastases[82], 64% were found to have originated from <strong>the</strong>head <strong>and</strong> neck region (including <strong>the</strong> skin), 11% fromdistant sites <strong>and</strong> 25% from an unknown primary. Of<strong>the</strong> distant sites, lung, kidney <strong>and</strong> breast accounted formore than four-fifths (Table 5.4); only four cases werefrom <strong>the</strong> prostate, but it is perhaps under-recognised[195]. Metastases to <strong>the</strong> subm<strong>and</strong>ibular gl<strong>and</strong>s are lesscommon than to <strong>the</strong> parotids, but are more likely to befrom distant sites [226].Microscopically, metastases in <strong>the</strong> salivary gl<strong>and</strong>scan resemble almost any primary tumour, so that forexample, mammary duct carcinoma is morphologicallyidentical (but immunohistochemically different) tosalivary duct carcinoma (see Sect. 5.9.9). Similarly, renalcell carcinoma is part <strong>of</strong> <strong>the</strong> differential diagnosis<strong>of</strong> any clear cell tumour <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s, <strong>and</strong> examples<strong>of</strong> prostate carcinoma have been mistaken foracinic cell carcinoma [195]. Immunohistochemistry is<strong>of</strong> some value, <strong>and</strong> can identify prostate <strong>and</strong> thyroidprimaries <strong>and</strong> melanoma with a reasonable degree <strong>of</strong>accuracy. Unlike most primary malignant salivary tumours,renal cell carcinomas are usually negative withcytokeratin 7; in contrast, CD10 stains most kidneycarcinomas, but is only positive in salivary tumourswith myoepi<strong>the</strong>lial differentiation. However, <strong>the</strong> possibility<strong>of</strong> metastasis is still best confirmed or excludedby imaging techniques <strong>of</strong> <strong>the</strong> kidneys.


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 1615.10 HybridCarcinomaHybrid tumours are composed <strong>of</strong> two different types <strong>of</strong>tumour, each <strong>of</strong> which conforms to an exactly definedcategory <strong>of</strong> tumour. They are rare, comprising


162 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5.13.2 PredominantlyEpi<strong>the</strong>lial Malignancies5Major salivary gl<strong>and</strong> carcinomas with acinar/ductal epi<strong>the</strong>lialdifferentiation include mucoepidermoid carcinoma(MEC), acinic cell carcinoma (AcCC) <strong>and</strong> salivaryduct carcinoma (SDC). Gene expression pr<strong>of</strong>iles havebeen determined in all three using Clontech’s cDNAarrays including 1,176 cancer-related genes [123]. Onlyfive such genes are overexpressed by all <strong>the</strong>se carcinomatypes including fibronectin (FN1), tissue metalloproteinaseinhibitor-1 (TIMP1), biglycan (BGN), tenascin C(HXB), <strong>and</strong> insulin-like growth-factor binding protein-5 (IGFBP5). Sixteen genes, i.e. KIAA0137/TLK1, VRK2,ADSL, CREBBP, PSM/FOLH, PIK3R1, PRKACB, BAG1,SMAD4/MADH4, TRAM, LAMA4, AKAP1, MAPK13,ATP5J, ATIC, <strong>and</strong> EPS15, are underexpressed in all <strong>of</strong><strong>the</strong>m.Average-linkage hierarchical clustering indicatesgenes that are significantly differently expressed among<strong>the</strong>se carcinoma types. They were identified using significanceanalysis <strong>of</strong> microarrays (SAM). In hierarchicalclustering, low-grade <strong>and</strong> high-grade MECs clusterclosely toge<strong>the</strong>r, <strong>and</strong> separate from closely clusteredSDC [123]. A hierarchical clustering <strong>of</strong> SDC <strong>and</strong> ACCshows that each entity clusters toge<strong>the</strong>r, <strong>and</strong> separatesfrom <strong>the</strong> o<strong>the</strong>r entity. In SAM, 27 genes are significantlydifferently expressed between MEC <strong>and</strong> SDC [123].Five genes, i.e. MMP11, DAP12, KIAA0324, FASN <strong>and</strong>CASP10 are overexpressed by SDC, while eight genes,including IL-6 <strong>and</strong> KRT14 are overexpressed by MEC.Ano<strong>the</strong>r 14 genes that are mainly involved in DNAmodification, such as NME4, NTHL1, RBBP4, HMG17<strong>and</strong> NDP52, are underexpressed by MEC. QuantitativeRT-PCR confirms results including overexpression <strong>of</strong>FN1 <strong>and</strong> TIMP1, underexpression <strong>of</strong> PSM/FOLH1 <strong>and</strong>MADH4/SMAD4, as well as <strong>the</strong> difference <strong>of</strong> expressionpr<strong>of</strong>iles <strong>of</strong> IL-6, CASP10 <strong>and</strong> KRT14 between SDC <strong>and</strong>MEC. Immunohistochemistry indicates distinct expression<strong>of</strong> cytokeratin 14 in MEC, with no expression inAcCC <strong>and</strong> SDC [123].Fur<strong>the</strong>rmore, major differences between predominantlymyoepi<strong>the</strong>lial <strong>and</strong> predominantly epi<strong>the</strong>lial carcinomasalso exist. In MEC <strong>and</strong> AdCC, <strong>the</strong> expression<strong>of</strong> important effector molecules such as erbB-2, erbB-3, epidermal growth factor receptor <strong>and</strong> transforminggrowth factor- is largely dissimilar [80].The small number <strong>of</strong> similarly deregulated genesin <strong>the</strong>se major types <strong>of</strong> salivary gl<strong>and</strong> carcinoma suggestsheterogenic mechanisms <strong>of</strong> tumorigenesis. Thisis consistent with <strong>the</strong> great histopathological diversityamong carcinomas <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s [171]. Diversityis also implicated by differences in gene expressionamong tumour types in hierarchical clustering. Thesmall number <strong>of</strong> genes jointly overexpressed by majorFig. 5.55. Lymphoepi<strong>the</strong>lial sialoadenitis (LESA): lymphoepi<strong>the</strong>liallesions are cohesive aggregates <strong>of</strong> epi<strong>the</strong>lial cells with hyalinisedbasal membrane-like material infiltrated by lymphocytessalivary gl<strong>and</strong> carcinomas with epi<strong>the</strong>lial differentiationrelate to cell adhesion, motility <strong>and</strong> cell shape (FN1,BGN, HXB), cancer growth, metastasis, tumour angiogenesis<strong>and</strong> apoptosis (TIMP-1, IGFBP-5). The jointlyunderexpressed genes include cell-cycle proteins, proteins<strong>of</strong> signal transduction <strong>and</strong> translation, <strong>and</strong> extracellularmatrix <strong>and</strong> membrane proteins. Interestingly,<strong>the</strong> jointly underexpressed CREBBP <strong>and</strong> MADH4/SMAD4 are intimately involved in <strong>the</strong> TGF-β transcriptionpathway <strong>of</strong> growth inhibition, <strong>and</strong> MADH4/SMAD4 has been shown to be deleted or mutated inhalf <strong>of</strong> pancreatic carcinomas [178]. Thus, overexpression<strong>of</strong> TIMP1, PLAT <strong>and</strong> SFN [38, 178], <strong>and</strong> underexpression<strong>of</strong> MADH4/SMAD4 are shared by carcinomas<strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s <strong>and</strong> <strong>the</strong> pancreas, but not witho<strong>the</strong>r exocrine carcinomas. In <strong>the</strong> salivary gl<strong>and</strong>s, coordinatedloss <strong>of</strong> SMAD4 <strong>and</strong> CREBBP functions couldimpair growth control <strong>and</strong> promote oncogenic transformation.5.14 Benign <strong>and</strong> MalignantLymphoid Infiltrates5.14.1 Non-AutoimmuneLymphoid InfiltratesThese can be considered in four groups – obstructive,infective, various non-infective inflammatory processes,<strong>and</strong> associated with epi<strong>the</strong>lial tumours, especiallycarcinomas (see Sect. 5.9).


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 163Table 5.5. Overview <strong>of</strong> autoimmune <strong>and</strong> neoplastic salivary lymphoid proliferations, adapted from Quintana et al. [165]BenignBorderlineLow-grade lymphomaLESA (lymphoepi<strong>the</strong>lial sialadenitis), non-clonal(Histological or clonal evidence <strong>of</strong> neoplasia, but unlikely to disseminate): LESA,clonal; LESA with halos <strong>of</strong> marginal zone B-cellsPotential for spread to nodes <strong>and</strong> less <strong>of</strong>ten, systemically: low-grade marginal zoneB-cell lymphoma <strong>of</strong> mucosa-associated lymphoid tissue (MALT lymphoma)– confluent proliferation <strong>of</strong> marginal zone B-cellsLow-grade marginal zone B-cell lymphoma <strong>of</strong> mucosa-associated lymphoid tissue(MALT lymphoma) with plasmacytic differentiation5.14.2 Benign AutoimmuneLymphoid InfiltratesThe most frequent form <strong>of</strong> autoimmune sialadenitis goesunder several synonyms, such as Mikulicz sialadenitis/disease, myoepi<strong>the</strong>lial sialadenitis (MESA) <strong>and</strong> benignlymphoepi<strong>the</strong>lial lesion, none <strong>of</strong> which is satisfactory[194]. In 1999, Harris introduced <strong>the</strong> much more accurateterm, lymphoepi<strong>the</strong>lial sialadenitis (LESA) [91], <strong>and</strong>this has begun to gain general acceptance, <strong>and</strong> will beused here. Sjögren’s syndrome is not a pathological term,but a clinical combination <strong>of</strong> dry eyes <strong>and</strong> mouth dueto autoimmune infiltrates <strong>of</strong> <strong>the</strong> lacrimal <strong>and</strong> salivarygl<strong>and</strong>s. It is <strong>of</strong>ten associated with o<strong>the</strong>r autoimmune orconnective tissue diseases, particularly rheumatoid arthritis,but also for example scleroderma, systemic lupusery<strong>the</strong>matosus, Hashimoto’s thyroiditis <strong>and</strong> chronicactive hepatitis. Most patients with Sjögren’s syndromedevelop LESA, but not so <strong>the</strong> reverse, as up to 50% <strong>of</strong>patients with LESA do not develop <strong>the</strong> clinical features<strong>of</strong> Sjögren’s syndrome [81].Lymphoepi<strong>the</strong>lial sialadenitis is considered to be anautoimmune disease [40, 77] <strong>of</strong> unknown aetiology. Severalviruses have been implicated [76, 88], but <strong>the</strong>y actprobably only as c<strong>of</strong>actors.About 80% <strong>of</strong> patients with LESA are female, with amean age at presentation <strong>of</strong> 55 years (range 1 to 86). Theparotids are involved in over 80% <strong>of</strong> cases (20% bilaterally),<strong>the</strong> subm<strong>and</strong>ibular gl<strong>and</strong>s in 11%, usually in combinationwith <strong>the</strong> parotids [40], <strong>and</strong> o<strong>the</strong>r sites in <strong>the</strong>head <strong>and</strong> neck in 6% [40, 81]. Tumour-like lesions <strong>of</strong> <strong>the</strong>minor gl<strong>and</strong>s are rare, but in contrast, subclinical focalperiductal <strong>and</strong> periacinar lymphoplasmacytic infiltratesare frequently seen in <strong>the</strong> labial salivary gl<strong>and</strong>s inSjögren’s syndrome, <strong>and</strong> a semi-quantitative assessment<strong>of</strong> <strong>the</strong> amount <strong>of</strong> inflammation in a lip biopsy may beuseful as part <strong>of</strong> <strong>the</strong> investigation <strong>of</strong> patients with a drymouth [41].In <strong>the</strong> earliest stages <strong>of</strong> LESA salivary ducts are dilated<strong>and</strong> surrounded by a lymphoid infiltrate withgerminal centres. B-cells appear to concentrate around<strong>the</strong> ducts <strong>and</strong> focally infiltrate <strong>the</strong> epi<strong>the</strong>lium, unlikein many non-autoimmune chronic inflammatory infiltrates,where lymphocytic invasion <strong>of</strong> <strong>the</strong> ducts isless marked. In LESA many B-cells are <strong>of</strong> monocytoidor marginal zone type [91]. They are slightly largerthan small lymphocytes <strong>of</strong> <strong>the</strong> mantle zone, <strong>and</strong> arecharacterised by nuclei with irregular outlines somewhatresembling centrocytes [105]. Plasma cells <strong>and</strong> T-lymphocytes are also present. In time, <strong>the</strong> ducts condense,with partial or complete loss <strong>of</strong> <strong>the</strong>ir lumina, t<strong>of</strong>orm lymphoepi<strong>the</strong>lial lesions (LELs) [105, 172, 194];<strong>the</strong>se were previously inaccurately called epimyoepi<strong>the</strong>lialisl<strong>and</strong>s <strong>and</strong> similarly, <strong>the</strong> old usage <strong>of</strong> LEL torefer to <strong>the</strong> whole lesion should be discontinued [91].LELs consist <strong>of</strong> cohesive aggregates <strong>of</strong> epi<strong>the</strong>lium withhyalinised basal lamina material containing variablenumbers <strong>of</strong> B-cells (Fig. 5.55). Myoepi<strong>the</strong>lial cells arepresent as well, but are <strong>of</strong>ten relatively inconspicuous[42]. As <strong>the</strong> disease progresses <strong>the</strong> acini become atrophied<strong>and</strong> are <strong>the</strong>n totally replaced by lymphoid tissueleading to clinical enlargement <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s.Monoclonality by PCR can be demonstrated in over40% <strong>of</strong> patients with LESA [165], but this alone is probablyinsufficient for a diagnosis <strong>of</strong> lymphoma, <strong>and</strong>stronger evidence is required from <strong>the</strong> demonstration<strong>of</strong> monoclonality by immunohistochemistry or flowcytometry [107].5.14.3 MalignantLymphomaICD-O:9590/3Overall, extranodal <strong>and</strong> nodal lymphomas represented16.3% <strong>of</strong> all malignant tumours <strong>of</strong> <strong>the</strong> major salivarygl<strong>and</strong>s at <strong>the</strong> AFIP from 1985 to 1995 [62]. Almost allextranodal lymphomas are marginal zone B-cell lymphoma(MALT lymphomas), which is <strong>the</strong> preferred terminology<strong>of</strong> <strong>the</strong> current WHO classification <strong>of</strong> lymphomas[108].


164 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5Mucosa-associated lymphoid tissue lymphomas usuallypresent clinically as parotid enlargement, sometimesbilateral [217]. There is <strong>of</strong>ten a history <strong>of</strong> Sjögren’ssyndrome, but not always. Cases have been reported inpost-transplant patients [101]. General lymphadenopathy<strong>and</strong> bone marrow involvement are unusual in MALTlymphoma, <strong>and</strong> such a presentation favours a diagnosis<strong>of</strong> nodal lymphoma.The histopathology <strong>of</strong> MALT lymphoma is intimatelylinked with that <strong>of</strong> LESA from which it develops– <strong>the</strong> risk <strong>of</strong> lymphoma in LESA has been estimatedat approximately 4–7% [91]. Histological criteriaalone cannot identify exactly when a clonal B-cell populationemerges in LESA, <strong>and</strong> in practice <strong>the</strong> processcan be considered not so much a sharp change fromone (benign) entity to ano<strong>the</strong>r (malignant) one, butra<strong>the</strong>r as a spectrum <strong>of</strong> lymphoma gradually evolvingfrom a purely inflammatory process (Table 5.5) [165].It can be considered that MALT lymphoma begins asan antigen-driven lymphoid proliferation that progressesfirst to monoclonality <strong>and</strong> <strong>the</strong>n, with <strong>the</strong> acquisition<strong>of</strong> secondary genetic changes, to MALT lymphoma[105].The microscopic picture evolves with time [105]; <strong>the</strong>earliest morphologically recognisable feature <strong>of</strong> malignancyis <strong>the</strong> proliferation <strong>of</strong> marginal zone B-cells t<strong>of</strong>orm a distinct halo-like zone around <strong>the</strong> LELs <strong>of</strong> LESA.As <strong>the</strong> lymphoma evolves in a background <strong>of</strong> LESA marginalzones B cells exp<strong>and</strong>, displace <strong>and</strong> <strong>the</strong>n replace <strong>the</strong>follicles. Alternatively, <strong>the</strong>y may colonise <strong>the</strong> germinalcentres assuming a follicular-like architecture (pseud<strong>of</strong>ollicles)[106]. In addition, <strong>the</strong>re may be foci <strong>of</strong> sclerosis,<strong>and</strong> infiltration by epi<strong>the</strong>lioid histiocytes, which canform small granulomas.Mucosa-associated lymphoid tissue lymphoma restrictedto <strong>the</strong> salivary gl<strong>and</strong>s is a relatively indolentdisease that is <strong>of</strong>ten curable with local treatment. Prognosisremains favourable even in <strong>the</strong> presence <strong>of</strong> o<strong>the</strong>rextranodal sites, including bone marrow [108].O<strong>the</strong>r primary non-MALT extranodal salivary lymphomasare rare, <strong>and</strong> are mainly T-cell neoplasms[29].Nodal non-Hodgkin’s lymphomas can involve <strong>the</strong>intra-salivary <strong>and</strong> adjacent lymph nodes, <strong>and</strong> <strong>the</strong>yshould be classified using <strong>the</strong> appropriate scheme [104,108].5.15 O<strong>the</strong>r TumoursA variety <strong>of</strong> s<strong>of</strong>t tissue <strong>and</strong> o<strong>the</strong>r non-salivary neoplasmsmay rarely present as tumours <strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong>s. These include solitary fibrous tumour, granularcell tumour, follicular dendritic cell sarcoma, inflammatorypseudotumour (inflammatory my<strong>of</strong>ibroblastictumour), primary malignant melanoma, primitive neuroectodermaltumour (PNET) <strong>and</strong> teratoma.5.16 Unclassified TumoursThe revised WHO classification defines this group asbenign or malignant tumours that cannot be placed inany <strong>of</strong> <strong>the</strong> categories [171]. This designation may be unavoidableif only a small quantity <strong>of</strong> tissue is availablefor study.References1. Albeck H, Bentzen J, Ockelmann HH, Nielsen NH, Bretlau P,Hansen HS (1993) Familial clusters <strong>of</strong> nasopharyngeal carcinoma<strong>and</strong> salivary gl<strong>and</strong> carcinomas in Greenl<strong>and</strong> natives.Cancer 72:196–2002. Alós LL, Cardesa A, Bombí JA, Mall<strong>of</strong>ré C, Cuchi A, TraserraJ (1996) Myoepi<strong>the</strong>lial tumors <strong>of</strong> salivary gl<strong>and</strong>s: a clinicopathologic,immunohistochemical <strong>and</strong> flow-cytometricstudy. Semin Diagn Pathol 13:138–1473. Altini M, Coleman H, Kienkle F (1997) Intra-vascular tumourin pleomorphic adenoma – a report <strong>of</strong> four cases. Histopathology31:55–594. Angeles-Angeles A, Caballero-Mendoza E, Tapia-Rangel B,Cortés-González R, Larriva-Sahd J (1998) Adenocarcinomagigante de células acinares de tipo quístico papilar en parótida.Rev Invest Clin 50:245–2485. Araújo VC de, Loducca SVL, Sobral APV, Kowalski AL,Soares F, Araújo NS de (2002) Salivary duct carcinoma: cytokeratin14 as a marker <strong>of</strong> in situ intraductal growth. Histopathology41:244–2496. Auclair P (1994) Tumor-associated lymphoid proliferationin <strong>the</strong> parotid gl<strong>and</strong>: a potential diagnostic pitfall. Oral SurgOral Med Oral Pathol 77:19–267. Auclair PL, Ellis GL (1996) Atypical features in salivarygl<strong>and</strong> mixed tumors: <strong>the</strong>ir relationship to malignant transformation.Mod Pathol 9:652–6578. Barbareschi M, Pecciarini L, Cangi MG, Macrì E, Rizzo A,Viale G, Doglioni C (2001) p63, a p53 homologue, is a selectivenuclear marker <strong>of</strong> myoepi<strong>the</strong>lial cells <strong>of</strong> <strong>the</strong> humanbreast. Am J Surg Pathol 25:1054–10608a. Barnes L, Eveson JW, Reichart P, Sidransky D, (eds) (2005)WHO Classification <strong>of</strong> Tumours. <strong>Pathology</strong> <strong>and</strong> Genetics.<strong>Head</strong> <strong>and</strong> <strong>Neck</strong> Tumours. Chapter 5. Salivary gl<strong>and</strong>s. IARCPress Lyon9. Barnes L, Rao U, Krause J, Contis L, Schwartz A, ScalamognaP (1994) Salivary duct carcinoma. I. A clinicopathologicevaluation <strong>and</strong> DNA image analysis <strong>of</strong> 13 cases with review<strong>of</strong> <strong>the</strong> literature. Oral Surg Oral Med Oral Pathol 78:64–7310. Batsakis JG (1979) Tumors <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck: clinical <strong>and</strong>pathological considerations, 2nd edn. Williams & Wilkins,Baltimore, p 11611. Batsakis JG (1996) Sclerosing polycystic adenosis: a newlyrecognized salivary gl<strong>and</strong> lesion, a form <strong>of</strong> chronic sialadenitis.Adv Anat Pathol 3:298–30412. Batsakis JG, El-Naggar AK (1990) Sebaceous lesions <strong>of</strong> salivarygl<strong>and</strong>s <strong>and</strong> oral cavity. Ann Otol Rhinol Laryngol99:416–41813. Batsakis JG, El-Naggar AK, Luna MA (1992) Epi<strong>the</strong>lial-myoepi<strong>the</strong>lialcarcinoma <strong>of</strong> salivary gl<strong>and</strong>s. Ann Otol RhinolLaryngol 101:540–54214. Batsakis JG, Frankenthaler R (1992) Embryoma (sialoblastoma)<strong>of</strong> salivary gl<strong>and</strong>s. Ann Otol Rhinol Laryngol 101:958–960


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 16515. Batsakis JG, Luna MA (1991) Undifferentiated carcinomas <strong>of</strong>salivary gl<strong>and</strong>s. Ann Otol Rhinol Laryngol 100:82–8416. Batsakis JG, Luna MA (1991) Basaloid salivary carcinomas.Ann Otol Rhinol Laryngol 100:785–78717. Batsakis JG, Manning JT (1987) Necrotizing sialometaplasia<strong>of</strong> major salivary gl<strong>and</strong>s. J Laryngol Otol 101:962–96618. Bergensen PJ, Kennedy PJ, Kneale KL (1987) Metastatic tumours<strong>of</strong> <strong>the</strong> parotid region. Aust NZ J Surg 57:23–2619. Br<strong>and</strong>wein-Gensler M, Hille J, Wang BY, Urken M, GordonR, Wang LJ, Simpson JR, Simpson RH, Gnepp DR (2004)Low-grade salivary duct carcinoma: description <strong>of</strong> 16 cases.Am J Surg Pathol 28:1040–104420. Br<strong>and</strong>wein MS, Huvos AG (1991) Oncocytic tumors <strong>of</strong> majorsalivary gl<strong>and</strong>s. A study <strong>of</strong> 68 cases with follow-up <strong>of</strong> 44 patients.Am J Surg Pathol 15:514–52821. Br<strong>and</strong>wein M, Huvos AG, Dardick I, Thomas MG, Theise ND(1996) Noninvasive <strong>and</strong> minimally invasive carcinoma exmixed tumor. A clinicopathologic <strong>and</strong> ploidy study <strong>of</strong> 12 patientswith major salivary tumors <strong>of</strong> low malignant potential.Oral Surg Oral Med Oral Pathol 81:665–66422. Br<strong>and</strong>wein MS, Ivanov K, Wallace DI, Hille JJ, Wang B, FahmyA, Bodian C, Urken ML, Gnepp DR, Huvos A, LumermanH, Mills SE (2001) Mucoepidermoid carcinoma: a clinicopathologicstudy <strong>of</strong> 80 patients with special reference tohistological grading. Am J Surg Pathol 25:835–84523. Br<strong>and</strong>wein MS, Jagirdar J, Patil J, Biller H, Kaneko M (1990)Salivary duct carcinoma (cribriform salivary carcinoma <strong>of</strong>excretory ducts): a clinicopathologic <strong>and</strong> immunohistochemicalstudy <strong>of</strong> 12 cases. Cancer 65:2307–231424. Brannon RB, Fowler CB, Hartman KS (1991) Necrotizing sialometaplasia:a clinicopathologic study <strong>of</strong> sixty-nine cases<strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Oral Surg Oral Med Oral Path72:317–32525. Buchner A, Merrell PW, Carpenter WM, Leider AS (1991)Adenomatoid hyperplasia <strong>of</strong> minor salivary gl<strong>and</strong>s. OralSurg Oral Med Oral Pathol 71:583–58726. Castle JT, Thompson LDR, Frommelt RA, Wenig BM, KesslerHP (1996) Polymorphous low-grade adenocarcinoma: a clinicopathologicstudy <strong>of</strong> 164 cases. Cancer 86:207–21927. Chan JKC, Hui PK, Ng CS, Yuen NWF, Kung ITM, Gwi E(1989) Epi<strong>the</strong>lioid haemangioma (angiolymphoid hyperplasiawith eosinophilia) <strong>and</strong> Kimura’s disease in Chinese. Histopathology15:557–57428. Chan JKC, Tang SK, Tsang WYW, Lee KC, Batsakis JG (1996)Histologic changes induced by fine-needle aspiration. AdvAnat Pathol 3:71–9029. Chan JKC, Tsang WYW, Hui P-K, Ng C-S, Sin V-C, KhanSM, Siu LLP (1997) T-<strong>and</strong> T/Natural killer-cell lymphomas<strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>: a clinicopathologic, immunohistochemical<strong>and</strong> molecular study <strong>of</strong> six cases. Hum Pathol28:238–24530. Chau MN, Radden BG (1989) A clinico-pathological study <strong>of</strong>53 intra-oral pleomorphic adenomas. Int J Oral Maxill<strong>of</strong>acSurg 18:158–16231. Chetty R (2000) Intercalated duct hyperplasia: possible relationshipto epi<strong>the</strong>lial-myoepi<strong>the</strong>lial carcinoma <strong>and</strong> hybridtumours <strong>of</strong> salivary gl<strong>and</strong>. Histopathology 37:260–26332. Cheuk W, Chan JK, Ngan RK (1999) Dedifferentiation inadenoid cystic carcinoma <strong>of</strong> salivary gl<strong>and</strong>: an uncommoncomplication associated with an accelerated clinical course.Am J Surg Pathol 23:465–47233. Chieng DC, Paulino AF (2002) Cytology <strong>of</strong> myoepi<strong>the</strong>lialcarcinoma <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>: a study <strong>of</strong> four cases. Cancer(Cancer Cytopathol) 96:32–3634. Cho KJ, El-Naggar AK, Ordóñez NG, Luna MA, Austin J,Batsakis JG (1995) Epi<strong>the</strong>lial-myoepi<strong>the</strong>lial carcinoma <strong>of</strong> salivarygl<strong>and</strong>s: a clinicopathologic, DNA flow cytometric, <strong>and</strong>immunohistochemical study <strong>of</strong> Ki-67 <strong>and</strong> HER-2/neu oncogene.Am J Clin Pathol 103:432–43735. Claros P, Dominte G, Claros Jr A, Castillo M, Cardesa A,Claros A (2002) Parotid gl<strong>and</strong> mucoepidermoid carcinomain a 4 year old child. Int J Pedriatr Otorhinolaryngol 63:67–7236. Colmenero Ruiz C, Patrón Romero M, Martín Pérez M (1993)Salivary duct carcinoma: a report <strong>of</strong> nine cases. J Oral Maxill<strong>of</strong>acSurg 51:641–64637. Croitoru CM, Suarez PA, Luna MA (1999) Hybrid carcinoma<strong>of</strong> salivary gl<strong>and</strong>s: report <strong>of</strong> 4 cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.Arch Pathol Lab Med 123:698–70238. Crnogorac-Jurcevic T, Efthimiou E, Nielsen T, Loader J, TerrisB, Stamp G, Baron A, Scarpa A, Lemoine NR (2002) Expressionpr<strong>of</strong>iling <strong>of</strong> microdissected pancreatic adenocarcinomas.Oncogene 21:4587–459439. Daley TD (1984) The canalicular adenoma: considerationson differential diagnosis <strong>and</strong> treatment. J Oral Maxill<strong>of</strong>acSurg 42:728–73040. Daniels TE (1991) Benign lymphoepi<strong>the</strong>lial lesion <strong>and</strong>Sjögren’s syndrome. In: Ellis GL, Auclair PL, Gnepp DR (eds)Surgical pathology <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. Saunders, Philadelphia,pp 83–10641. Daniels TE, Fox PS (1992) Salivary <strong>and</strong> oral components <strong>of</strong>Sjögren’s syndrome. Rheum Dis Clin N Am 18:571–58942. Dardick I, van Nostr<strong>and</strong> P, Rippstein P, Skimming L, HoppeD, Dairkee SH (1988) Characterization <strong>of</strong> epimyoepi<strong>the</strong>lialisl<strong>and</strong>s in benign lymphoepi<strong>the</strong>lial lesions <strong>of</strong> major salivarygl<strong>and</strong>: an immunohistochemical <strong>and</strong> ultrastructural study.<strong>Head</strong> <strong>Neck</strong> Surg 10:168–17843. Dardick I, Thomas MJ, Nostr<strong>and</strong> AWP van (1989) Myoepi<strong>the</strong>lioma– new concepts <strong>of</strong> histology <strong>and</strong> classification: alight <strong>and</strong> electron microscopic study. Ultrastruct Pathol13:187–22444. Deb RA, Desai SB, Amonkar PP, Aiyer PM, Borges AM(1998) Primary primitive neuroectodermal tumour <strong>of</strong> <strong>the</strong>parotid gl<strong>and</strong>. Histopathology 33:375–37845. Dehner LP, Valbuena L, Perez-Atayde A, Reddick RL, AskinFB, Rosai J (1984) Salivary gl<strong>and</strong> anlage tumor (“congenitalpleomorphic adenoma”): a clinicopathologic, immunohistochemical<strong>and</strong> ultrastructural study <strong>of</strong> nine cases. Am J SurgPathol 18:25–3646. Delgado R, Klimstra D, Albores-Saavedra J (1996) Low gradesalivary duct carcinoma: a distinctive variant with a lowgrade histology <strong>and</strong> a predominant intraductal growth pattern.Cancer 78:958–96747. Delgado R, Vuitch F, Albores-Saavedra J (1993) Salivary ductcarcinoma. Cancer 72:1503–151248. Di Palma S (1994) Epi<strong>the</strong>lial-myoepi<strong>the</strong>lial carcinoma withco-existing multifocal intercalated duct hyperplasia <strong>of</strong> <strong>the</strong>parotid gl<strong>and</strong>. Histopathology 25:494–49649. Di Palma S (2003) Malignancy in pleomorphic adenoma(malignant mixed tumour) <strong>of</strong> salivary gl<strong>and</strong>s. Gale N, LuzarB (eds) Proceedings in head <strong>and</strong> neck pathology. Institute <strong>of</strong><strong>Pathology</strong> Faculty <strong>of</strong> Medicine, University <strong>of</strong> Lubljana, Lubljana4–6 September 2003, pp 92–9850. Di Palma S (2004) Case 23, slide seminar <strong>of</strong> head <strong>and</strong> neckpathology. II. Intercontinental Congress <strong>of</strong> <strong>Pathology</strong>, Iguassu,Brazil51. Di Palma S, Corletto V, Lavarino C, Birindelli S, Pilotti S(1999) Unilateral aneuploid dedifferentiated acinic cell carcinomaassociated with bilateral low grade diploid acinic cellcarcinoma <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>. Virchows Arch 434:361–36552. Di Palma S, Guzzo M (1993) Malignant myoepi<strong>the</strong>lioma <strong>of</strong>salivary gl<strong>and</strong>s: clinicopathological features <strong>of</strong> ten cases.Virchows Arch A Pathol Anat 423:389–39653. Di Palma S, Guzzo M (1998) Myoepi<strong>the</strong>lial carcinoma withpredominance <strong>of</strong> plasmacytoid cells arising in a pleomorphicadenoma <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>. Histopathology 33:48554. Di Palma S, Pilotti S, Rilke F (1991) Malignant myoepi<strong>the</strong>lioma<strong>of</strong> <strong>the</strong> parotid arising in a pleomorphic adenoma. Histopathology19:273–27555. Di Palma S, Simpson RHW, Skálová A, Michal M (1999)Metaplastic (infarcted) Warthin’s tumour <strong>of</strong> <strong>the</strong> parotidgl<strong>and</strong>: a possible consequence <strong>of</strong> fine needle aspiration biopsy.Histopathology 35:432–43856. Di Palma S, Skálová A (1999) Malignancy in pleomorphic adenoma.Rev Esp Patol 32:431–432


166 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo557. Di Palma S, Skalova A, Vanecek T, Simpson RHW, StarekI, Leivo I (2005) Non invasive intracapsular) carcinoma expleomorphic adenoma: recognition <strong>of</strong> focal carcinoma byHER-2/neu <strong>and</strong> MIB-1 immunohistochemistry. Histopathology46:144–15258. Donath K, Seifert G (1997) Sclerosing polycystic siadadenopathy.Pathologe 18:368–37359. Edwards PC, Bhuiya T, Kelsch RD (2003) C-kit expressionin <strong>the</strong> salivary gl<strong>and</strong> neoplasms adenoid cystic carcinoma,polymorphous low-grade adenocarcinoma, <strong>and</strong> monomorphicadenoma. Oral Surg Oral Med Oral Pathol Oral RadiolEndod 95:686–69360. Elliott JN, Oertel YC (1990) Lymphoepi<strong>the</strong>lial cysts <strong>of</strong> <strong>the</strong>salivary gl<strong>and</strong>s: histologic <strong>and</strong> cytologic features. Am J ClinPathol 93:39–4361. Ellis GL (1988) “Clear cell” oncocytoma <strong>of</strong> salivary gl<strong>and</strong>.Hum Pathol 19:862–86762. Ellis GL, Auclair PL (1996) Malignant epi<strong>the</strong>lial tumors. In:Atlas <strong>of</strong> tumor pathology, 3rd series, fascicle 17: tumors <strong>of</strong><strong>the</strong> salivary gl<strong>and</strong>s. AFIP, Washington DC63. Evans HL, Luna MA (2000) Polymorphous low-grade adenocarcinoma:a study <strong>of</strong> 40 cases with long-term follow up <strong>and</strong>evaluation <strong>of</strong> <strong>the</strong> importance <strong>of</strong> papillary areas. Am J SurgPathol 24:1319–132864. Eveson JW (1992) Troublesome tumours 2: borderline tumours<strong>of</strong> salivary gl<strong>and</strong>s. J Clin Pathol 45:369–37765. Eveson JW (1999) Oncocytic <strong>and</strong> squamous differentiationin salivary gl<strong>and</strong>s <strong>and</strong> tumors. Rev Esp Patol 32:433–43466. Eveson JW, Cawson RA (1989) Infarcted (‘infected’) adenolymphomas.A clinicopathological study <strong>of</strong> 20 cases. ClinOtolaryngol 14:205–21067. Feind CR (1972) The head <strong>and</strong> neck. In: Haagensen CD,Feind CR, Herter FP (eds) The lymphatics in cancer. Saunders,Philadelphia, pp 63–6468. Felix A, Rosa-Santos J, Mendoca ME, Torrinha F, Soares J(2002) Intracapsular carcinoma ex pleomorphic adenoma.Report <strong>of</strong> a case with unusual metastatic behaviour. Oral Oncol38:107–11069. Ferlito A, Gale N, Hvala H (1981) Laryngeal salivary ductcarcinoma: a light <strong>and</strong> electron microscopic study. J LaryngolOtol 95:731–73870. Ferreiro JA (1994) Immunohistochemistry <strong>of</strong> basal cell adenoma<strong>of</strong> <strong>the</strong> major salivary gl<strong>and</strong>s. Histopathology 24:539–54271. Fonseca I, Soares J (1993) Epi<strong>the</strong>lial-myoepi<strong>the</strong>lial carcinoma<strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. A study <strong>of</strong> 22 cases. Virchows ArchA Pathol Anat 422:389–39672. Fonseca I, Soares J (1996) Basal cell adenocarcinoma <strong>of</strong> minorsalivary <strong>and</strong> seromucinous gl<strong>and</strong>s <strong>of</strong> <strong>the</strong> head <strong>and</strong> neckregion. Semin Diagn Pathol 13:128–13773. Fonseca I, Felix A, Soares J (2000) Dedifferentiation in salivarygl<strong>and</strong> carcinomas. Am. J. Surg Pathol 24:469–47174. Foschini M, Scarpellini F, Gown AM, Eusebi V (2000) Differentialexpression <strong>of</strong> myoepi<strong>the</strong>lial markers in salivary,sweat <strong>and</strong> mammary gl<strong>and</strong>s. Int J Surg Pathol 8:29–3775. Foucar E, Rosai J, Dorfman R (1990) Sinus histiocytosis withmassive lymphadenopathy (Rosai-Dorfman disease): review<strong>of</strong> <strong>the</strong> entity. Semin Diagn Pathol 7:19–7376. Fox RI, Pearson G, Vaughan JH (1986) Detection <strong>of</strong> Epstein-Barr virus-associated antigens <strong>and</strong> DNA in salivary gl<strong>and</strong>biopsies from patients with Sjögren’s syndrome. J Immunol137:3162–316877. Fox RI, Kang HI (1992) Pathogenesis <strong>of</strong> Sjögren’s syndrome.Rheum Dis Clin N Am 18:517–53878. Francioso F, Carinci F, Tosi L, Scapoli L, Pezetti F, PasserellaE, Evangelisti R, Pastore A, Pelucchi S, Piattelli A, Rubini C,Fioroni M, Carinci P, Volinia S (2002) Identification <strong>of</strong> differentiallyexpressed genes in human salivary gl<strong>and</strong> tumors byDNA microarrays. Mol Cancer Ther 1:533–53879. Frierson HF, El-Naggar AK, Welsh JB, Sapinoso LM, Su, AI,Cheng J, Saku T, Moskaluk CA, Hampton GM (2002) Largescale molecular analysis identifies genes with altered expressionin salivary adenoid cystic carcinoma. Am J Surg Pathol161:1315–132380. Gibbons MD, Manne U, Carroll WR, Peters GE, Weiss HL,Grizzle WE (2001) Molecular differences in mucoepidermoidcarcinoma <strong>and</strong> adenoid cystic carcinoma <strong>of</strong> <strong>the</strong> majorsalivary gl<strong>and</strong>s. Laryngoscope 111:1373–137881. Gleeson MJ, Cawson RA, Bennett MH (1986) Benign lymphoepi<strong>the</strong>liallesion: a less than benign disease. Clin Otolaryngol11:47–5182. Gnepp DR (1991) Metastatic disease to <strong>the</strong> major salivarygl<strong>and</strong>s. In: Ellis GL, Auclair PL, Gnepp DR (eds) Surgicalpathology <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. Saunders, Philadelphia, pp560–56983. Gnepp DR (1993) Malignant mixed tumors <strong>of</strong> salivary gl<strong>and</strong>s:a review. Pathol Annu 28:279–32884. Gnepp DR (2003) Sclerosing polycystic adenosis <strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong>: a lesion that may be associated with dysplasia <strong>and</strong>carcinoma in situ. Adv Anat Pathol 10:218–22285. Gnepp DR, Brannon R (1984) Sebaceous neoplasms <strong>of</strong> salivarygl<strong>and</strong> origin: report <strong>of</strong> 21 cases. Cancer 53:2155–217086. Grenko R T, Gemryd P, Tytor M, Lundqvist PG, Boeryd B(1995) Salivary duct carcinoma. Histopathology 26:261–26687. Guzzo M, Di Palma S, Gr<strong>and</strong>i C, Molinari R (1997) Salivaryduct carcinoma: clinical characteristics <strong>and</strong> treatment strategies.<strong>Head</strong> <strong>Neck</strong> 19:126–13388. Haddad J, Deny P, Munz-Go<strong>the</strong>il C, Ambrosini JC, TrinchetJC, Pateron D, Mal F, Callard P, Beaugr<strong>and</strong> M (1992) Lymphocyticsialadenitis <strong>of</strong> Sjögren’s syndrome associated withchronic hepatitis C virus liver disease. Lancet 339:321–32389. Hamilton-Dutoit SJ, Therkildsen MH, Nielsen NH, Jensen H,Hansen JPH, Pallesen G (1991) Undifferentiated carcinoma<strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong> in Greenl<strong>and</strong>ic Eskimos: demonstration<strong>of</strong> Epstein-Barr virus DNA by in situ nucleic acid hybridization.Hum Pathol 22:811–81590. Harmse JL, Saleh HA, Odutoye T, Alsanjari NA, MountainRE (1997) Recurrent canalicular adenoma <strong>of</strong> <strong>the</strong> minor salivarygl<strong>and</strong>s in <strong>the</strong> upper lip. J Larygol Otol 111:985–98791. Harris NL (1999) Lymphoid proliferations <strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong>s. Am J Clin Pathol 111 [Suppl 1]:S94–S10392. Heikinheimo AK, Laine MA, Ritvos OV-P, Voutilainen RJ,Hogan BLM, Leivo I (1999) Bone morphogenetic protein-6 isa marker <strong>of</strong> serous acinar cell differentiation in normal <strong>and</strong>neoplastic human salivary gl<strong>and</strong>. Cancer Res 59:5815–582193. Hellquist HB, Karlsson MG, Nilsson C (1994) Salivary ductcarcinoma – a highly aggressive salivary gl<strong>and</strong> tumour withoverexpression <strong>of</strong> c-erbB-2. J Pathol 172:35–4494. Hellquist HB, Sundelin K, Di Bacco A, Tytor M, ManzottiM, Viale G (1997) Tumour growth fraction <strong>and</strong> apoptosis insalivary gl<strong>and</strong> acinic cell carcinomas. Prognostic implications<strong>of</strong> Ki-67 <strong>and</strong> bcl-2 expression <strong>and</strong> <strong>of</strong> in situ end labelling(TUNEL). J Pathol 181:323–32995. Henley JD, Geary WA, Jackson CL, Wu CD, Gnepp DR (1997)Dedifferentiated acinic cell carcinoma <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>: adistinct rarely described entity. Hum Pathol 28:869–87396. Henley JD, Seo IS, Dayan D, Gnepp DR (2000). Sarcomatoidsalivary duct carcinoma <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>. Hum Pathol31:208–21397. Henriksson G, Westrin KM, Carlsöö B, Silfverswärd C (1998)Recurrent primary pleomorphic adenomas <strong>of</strong> salivary gl<strong>and</strong>origin: intrasurgical rupture, histopathologic features, <strong>and</strong>pseudopodia. Cancer 82:617–62098. Herold J, Nicholson AG (1992) Fine needle aspiration cytologyin <strong>the</strong> diagnosis <strong>of</strong> amyloid in <strong>the</strong> subm<strong>and</strong>ibular gl<strong>and</strong>.Br J Oral Maxill<strong>of</strong>ac Surg 30:393–39499. Hoang MP, Callender DL, Sola-Gallego JJ, Huang Z, SneigeN, Luna MA, Batsakis JG, El-Naggar AK (2001) Molecular<strong>and</strong> biomarker analyses <strong>of</strong> salivary duct carcinomas: comparisonwith mammary duct carcinoma. Int J Oncol 19:865–871


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 167100. Honda K, Kashima K, Yokohama S, Nakayama I (2000)Clonal analysis <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lial component <strong>of</strong> Warthin’s tumor.Hum Pathol 31:1377–1380101. Hsi ED, Singleton TP, Swinnen L, Dunphy CH, Alkan S(2000) Mucosa-associated lymphoid tissue-type lymphomasoccurring in post-transplantation patients. Am J Surg Pathol24:100–106102. Hsueh C, Gonzalez-Crussi F (1992) Sialoblastoma: a case report<strong>and</strong> review <strong>of</strong> <strong>the</strong> literature on congenital epi<strong>the</strong>lial tumors<strong>of</strong> salivary gl<strong>and</strong> origin. Pediatr Pathol 12:205–214103. Hui KK, Batsakis JG, Luna MA, MacKay B, Byers RM (1986)Salivary duct adenocarcinoma: a high grade malignancy. JLaryngol Otol 100:105–114104. Isaacson PG (1996) Malignant lymphomas with a folliculargrowth pattern. Histopathology 28:487–495105. Isaacson PG, Norton AJ (1994) Malignant lymphoma <strong>of</strong> <strong>the</strong>salivary gl<strong>and</strong>s. In: Isaacson PG, Norton AG (eds) Extranodallymphomas. Churchill-Livingstone, Edinburgh, pp 67–83106. Isaacson PG, Wo<strong>the</strong>rspoon AC, Diss TC, Pan LX (1991) Follicularcolonization in B-cell lymphoma <strong>of</strong> MALT. Am J SurgPathol 15:819–828107. Jaffe ES (2002) Lymphoid lesions <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck: amodel <strong>of</strong> lymphocyte homing <strong>and</strong> lymphomagenesis. ModPathol 15:255–263108. Jaffe ES, Harris NL, Stein H, Vardiman JW (2001) WorldHealth Organization classification <strong>of</strong> tumours: tumours <strong>of</strong>haematopoietic <strong>and</strong> lymphoid tissues. Springer, Berlin HeidelbergNew York109. Jahan-Parwar B, Huberman RM, Donovan D (1999) Oncocyticmucoepidermoid carcinoma <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. AmJ Surg Pathol 23:523–529110. James GK, Pudek M, Berean KW, Diam<strong>and</strong>is EP, ArchibaldBL (1996) Salivary duct carcinoma secreting prostate-specificantigen. Am J Clin Pathol 106:242–247111. Jayakrishnan A, Elmalah I, Hussain K, Odell EW (2003) Basalcell adenocarcinoma in minor salivary gl<strong>and</strong>s. Histopathology42:610–614112. Jeng YM, Lin CY, Hsu HC (2000) Expression <strong>of</strong> <strong>the</strong> c-kit proteinis associated with certain subtypes <strong>of</strong> salivary gl<strong>and</strong> carcinoma.Cancer Lett 154:107–111113. Jensen JL (1991) Idiopathic diseases. In: EllisGL, Auclair PL,Gnepp DR.(eds) Surgical pathology <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s.Saunders, Philadelphia, pp 60–82114. Jones H, Moshtael F, Simpson RHW (1992) Immunoreactivity<strong>of</strong> smooth muscle actin in salivary gl<strong>and</strong> tumours: a comparisonwith S 100 protein. J Clin Pathol 45:938–940115. Kay PA, Roche PC, Olsen KD, Lewis JE (2001) Salivary ductcarcinoma: immunohistochemical analysis <strong>of</strong> <strong>and</strong>rogen receptor,prostate markers <strong>and</strong> HER-2/neu oncoprotein in 40cases. [Abstract]. Mod Pathol 14:150A116. Kemp BL, Batsakis JG, El-Naggar AK, Kotliar SN, Luna MA(1995) Terminal duct adenocarcinomas <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>.J Laryngol Otol 109:466–468117. Kokemueller H, Eckardt A, Brachvogel P, Hausamen JE(2004) Adenoid cystic carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck – a 20years experience. Int J Oral Maxill<strong>of</strong>ac Surg 33:25–31118. Kraemer BB, MacKay B, Batsakis JG (1983) Small cell carcinomas<strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>: a clinicopathologic study <strong>of</strong>three cases. Cancer 52:2115–2121119. Kratochvil FJ (1991) Canalicular adenoma <strong>and</strong> basal cell adenoma.In: Ellis GL, Auclair PL, Gnepp DR (eds) Surgical pathology<strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. Saunders, Philadelphia, pp202–224120. Krieken JHJM van (1993) Prostate marker immunoreactivityin salivary gl<strong>and</strong> neoplasms: a rare pitfall in immunohistochemistry.Am J Surg Pathol 17:410–414121. Krishnamurthy S, Lanier AP, Dohan P, Lanier JF, Henle W(1988) Salivary gl<strong>and</strong> cancer in Alaskan natives, 1966–1980.Hum Pathol 18:986–996; erratum 19:328122. Kumar RV, Kini L, Bhargava AK, Mukherjee G, HazarikhaD, Shenoy AM, Anantha N (1993) Salivary duct carcinoma. JSurg Oncol 54:193–198123. Leivo I, Jee, KJ, Heikinheimo K, Laine M, Ollila J, Nagy B,Knuutila S (2005) Characterization <strong>of</strong> gene expression in majortypes <strong>of</strong> salivary gl<strong>and</strong> carcinoma with epi<strong>the</strong>lial differentiation.Cancer Genet Cytogenet 156:104–113124. Lewis JE, Olsen KD, Sebo TJ (2001) Carcinoma ex pleomorphicadenoma: pathologic analysis <strong>of</strong> 73 cases. Hum Pathol32:596–604125. LiVolsi VA, Perzin KH (1977) Malignant mixed tumor arisingin salivary gl<strong>and</strong>: carcinomas arising in benign mixed tumor.A clinico-pathologic study. Cancer 39:2209–2230126. Li S, Baloch ZW, Tomaszewski JE, Livolsi VA (2000) Worrisomehistologic alteration following fine-needle aspiration <strong>of</strong>benign parotid lesions. Arch Pathol Lab Med 124:87–91127. Luna MA, Batsakis JG, Tortoledo ME, del Junco GW (1989)Carcinomas ex monomorphic adenoma <strong>of</strong> salivary gl<strong>and</strong>s. JLaryngol Otol 103:756–759128. Luna MA, Tortoledo ME, Allen M (1987) Salivary dermal analoguetumors arising in lymph nodes. Cancer 59:1165–1169129. Martinez-Madrigal F, Micheau C (1989) Histology <strong>of</strong> <strong>the</strong> majorsalivary gl<strong>and</strong>s. Am J Surg Pathol 13:879–899130. Maurizi M, Salvinelli F, Capelli A (1990) Monomorphic adenomas<strong>of</strong> <strong>the</strong> major salivary gl<strong>and</strong>s: clinicopathologicalstudy <strong>of</strong> 44 cases. J Laryngol Otol 104:790–796131. McCluggage G, Sloan J, Cameron S, Hamilton P, Toner P(1995) Basal cell adenocarcinoma <strong>of</strong> <strong>the</strong> subm<strong>and</strong>ibulargl<strong>and</strong>. Oral Surg Oral Med Oral Pathol Oral Radiol Endod79:342–350132. Michal M, Skálová A, Simpson RHW, Rychterová V, Leivo I(1996) Clear cell malignant myoepi<strong>the</strong>lioma <strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong>s. Histopathology 28:309–315133. Michal M, Skálová A, Simpson RHW, Leivo I, Ryška A,Stárek I (1997) Well differentiated acinic cell carcinoma <strong>of</strong>salivary gl<strong>and</strong>s associated with lymphoid rich stroma: clinicopathologic<strong>and</strong> immunohistochemical study using a proliferativemarker MIB1. Hum Pathol 28:595–600134. Michal M, Skálová A, Simpson RHW, Raslan WF, Čuřík R,Leivo I, Mukenšnábl P (1999) Cribriform adenocarcinoma <strong>of</strong><strong>the</strong> tongue: a hi<strong>the</strong>rto unrecognized type <strong>of</strong> adenocarcinomacharacteristically occurring in <strong>the</strong> tongue. Histopathology35:495–501135. Michal M, Skálová A, Hrabal P (2002) Micropapillary carcinoma<strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong> arising in mucinous cystoadenoma.Virchows Arch 437:465–468136. Michal M, Sokol L, Mukenšnábl P (1996) Salivary gl<strong>and</strong> anlagetumor. A case with widespread necrosis <strong>and</strong> large cystformation (congenital pleomorphic adenoma). <strong>Pathology</strong>28:128–130137. Mikulicz J von (1982) Über eine eigenartige symmetrischeErkrankung der Tränen- und Mundspeicheldrüsen. BeitrChir Festschr Theodor Billroth, pp 610–630138. Milchgrub S, Gnepp DR, Vuitch F, Delgado R, Albores-SaavedraJ (1994) Hyalinizing clear cell carcinoma <strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong>. Am J Surg Pathol 18:74–82139. Mima T, Shirasuna K, Kishino M, Matsuya T (1996) Basalcell adenocarcinoma <strong>of</strong> <strong>the</strong> sublingual gl<strong>and</strong>: report <strong>of</strong> a case.J Oral Maxill<strong>of</strong>ac Surg 54:1121–1123140. Moles MA, Avila IR, Archilla AR (1999) Dedifferentiationoccurring in adenoid cystic carcinoma <strong>of</strong> <strong>the</strong> tongue. OralSurg Oral Med Oral Pathol Oral Radiol Endod 88:177–180141. Moody AB, Avery CME, Harrison JD (1998) Dermoid cyst <strong>of</strong><strong>the</strong> parotid gl<strong>and</strong>. Int J Oral Maxill<strong>of</strong>ac Surg 27:461–462142. Moriki T, Ueta S, Takahashi T, Mitani M, Ichien M (2001)Salivary duct carcinoma: cytologic characteristics <strong>and</strong> application<strong>of</strong> <strong>and</strong>rogen receptor immunostaining for diagnosis.Cancer (Cancer Cytopathol) 93:344–350143. Moritani S, Kushima R, Sugihara H, Bamba M, KobayashiTK, Hattori T (2002) Availability <strong>of</strong> CD10 immunohistochemistryas a marker <strong>of</strong> breast myoepi<strong>the</strong>lial cells on paraffinsections. Mod Pathol 15:397–405144. Muller S, Barnes LE (1996) Basal cell adenocarcinoma <strong>of</strong> <strong>the</strong>salivary gl<strong>and</strong>s: report <strong>of</strong> seven cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.Cancer 78:2471–2477


168 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo5145. Murrah VA, Batsakis JG (1994) Salivary duct carcinoma.Ann Otol Rhinol Laryngol 103:244–247146. Nagao T, Gaffey TA, Kay PA, Unni KK, Nascimento AG, SeboTJ, Serizawa H, Minato H, Lewis JE (2003) Dedifferentiationin low-grade mucoepidermoid carcinoma <strong>of</strong> <strong>the</strong> parotidgl<strong>and</strong>. Hum Pathol 34:1068–1072147. Nagao T, Gaffey TA, Visscher DW, Kay PA, Minato H, SerizawaH (2004) Invasive micropapillary salivary duct carcinoma:a distinct histologic variant with biologic significance.Am J Surg Pathol 28:319–326148. Nagao T, Gaffey TA, Olsen KD, Serizawa H, Lewis JE (2004)Small cell carcinoma <strong>of</strong> <strong>the</strong> major salivary gl<strong>and</strong>s. Clinicopathologicstudy with emphasis on cytokeratin immunoreactivity<strong>and</strong> clinical outcome. Am J Surg Pathol 28:762–770149. Nagao K, Matsuzaki O, Saiga H, Sugano I, Shigematsu H,Kaneko T, Katoh T, Kitamura T (1981) Histopathologic studieson carcinoma in pleomorphic adenoma <strong>of</strong> <strong>the</strong> parotidgl<strong>and</strong>. Cancer 48:113–121150. Nagao T, Serizawa H, Iwaya K, Shimizu T, Sugano I, Ishida Y,Yamazaki K, Shimizu M, Itoh T, Konno A, Ebihara Y (2002)Keratocystoma <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>: a report <strong>of</strong> two cases <strong>of</strong>an unusual pathologic entity. Mod Pathol 15:1005–1010151. Nagao T, Sugano I, Ishida Y, Tajima Y, Matsuzaki O, KonnoA, Kondo Y, Nagao K (1998) Salivary gl<strong>and</strong> malignant myoepi<strong>the</strong>lioma:a clinicopathologic <strong>and</strong> immunohistochemicalstudy <strong>of</strong> ten cases. Cancer 83:1292–1299152. Nagao T, Sugano I, Ishida Y, Hasegawa M, Matsuzaki O,Konno A, Kondo Y, Nagao K (1998) Basal cell adenocarcinoma<strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s; comparison with basal cell adenomathrough assessment <strong>of</strong> cell proliferation, apoptosis, <strong>and</strong>expression <strong>of</strong> p53 <strong>and</strong> bcl-2. Cancer 82:439–447153. Nasser SM, Faquin WC, Dayal Y (2003) Expression <strong>of</strong> <strong>and</strong>rogen,estrogen, <strong>and</strong> progesterone receptor in salivary gl<strong>and</strong>tumours. Frequent expression <strong>of</strong> <strong>and</strong>rogen receptor in a subset<strong>of</strong> malignant salivary gl<strong>and</strong> tumours. Am J Clin Pathol119:801–806154. Nunes JF, Fonseca I, Soares J (1996) Helioid inclusions in dedifferentiatedacinic cell carcinoma <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>. UltrastructPathol 20:443–449155. Nyrop M (1994) Kimura’s disease: case report <strong>and</strong> brief review<strong>of</strong> <strong>the</strong> literature. J Laryngol Otol 108:1005–1007156. Ogawa I, Nishida T, Miyauchi M, Sato S, Takata T (2003)Dedifferentiated malignant myoepi<strong>the</strong>lioma <strong>of</strong> <strong>the</strong> parotidgl<strong>and</strong>. Pathol Int 53:704–709157. Ogawa Y, Kishino M, Atsumi Y, Kimoto M, Fukuda Y, IshidaT (2003) Plasmocytoid cells in salivary-gl<strong>and</strong> pleomorphicadenomas: evidence <strong>of</strong> luminal differentiation. VirchowsArch 443:625–634158. Palmer TJ, Gleeson MJ, Eveson JW, Cawson RA (1990) Oncocyticadenomas <strong>and</strong> oncocytic hyperplasia <strong>of</strong> salivarygl<strong>and</strong>s. A clinicopathological study <strong>of</strong> 26 cases. Histopathology16:487–493159. Parrett TJ, Prasad AR, Raslan WF, Kakar S, Lewis JE (2002)Long term <strong>and</strong> life-long follow-up in polymorphous lowgrade adenocarcinoma (Abstract). Mod Pathol 15:223A160. Pelkey TJ, Mills SE (1999) Histologic transformation <strong>of</strong> polymorphouslow-grade adenocarcinoma <strong>of</strong> salivary gl<strong>and</strong>. AmJ Clin Pathol 111:785–791161. Perzin KH, LiVolsi VA (1980) Acinic cell carcinoma arisingin ectopic salivary gl<strong>and</strong> tissue. Cancer 45:967–972162. Pesce C, Colacino R, Buffa P (1986) Duct carcinoma <strong>of</strong> <strong>the</strong> minorsalivary gl<strong>and</strong>s: a case report. J Laryngol Otol 100:611–613163. Pieterse AS, Seymore AE (1981) Parotid cysts: an analysis <strong>of</strong>16 cases <strong>and</strong> suggested classification. <strong>Pathology</strong> 13:225–234164. Prasad AR, Savera AT, Gown AM, Zarbo RJ (1999) The myoepi<strong>the</strong>lialimmunophenotype in 135 benign <strong>and</strong> malignantsalivary gl<strong>and</strong> tumors o<strong>the</strong>r than pleomorphic adenoma.Arch Pathol Lab Med 123:801–806165. Quintana PG, Kapadia SB, Bahler DW, Johnson JT, SwerdlowSH (1997) Salivary gl<strong>and</strong> lymphoid infiltrates associatedwith lymphoepi<strong>the</strong>lial lesions: a clinicopathologic, immunophenotypic,<strong>and</strong> genotypic study. Hum Pathol 28:850–861166. Reis-Filho JS, Simpson RHW, Fulford LG, Steppeler Y, MartinsA, Schmitt FC (2004) p63 distribution in normal salivarygl<strong>and</strong> <strong>and</strong> salivary gl<strong>and</strong> neoplasms (abstract). ModPathol 17:231A167. Russo G, Zegar C, Giordano A (2003) Advantages <strong>and</strong> limitations<strong>of</strong> microarray technology in human cancer. Oncogene22:6497–6507168. Savera AT, Sloman A, Huvos AG, Klimstra DS (2000) Myoepi<strong>the</strong>lialcarcinoma <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s: a clinicopathologicstudy <strong>of</strong> 25 patients. Am J Surg Pathol 24:761–774169. Scher RL, Feldman PS, Lambert PR (1991) Oncocytic malignancy<strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg105:868–876170. Sciubba JJ, Brannon RB (1982) Myoepi<strong>the</strong>lioma <strong>of</strong> salivarygl<strong>and</strong>s: report <strong>of</strong> 23 cases. Cancer 49:562–572171. Seifert G (1991) World Health Organization internationalhistological classification <strong>of</strong> tumours: histological typing <strong>of</strong>salivary gl<strong>and</strong> tumours, 2nd edn. Springer, Berlin HeidelbergNew York172. Seifert G (1992) Tumour-like lesions <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s:<strong>the</strong> new WHO classification. Pathol Res Pract 188:836–846173. Seifert G (1996) Mucoepidermoid carcinoma in a salivaryduct cyst <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>: contribution to <strong>the</strong> development<strong>of</strong> tumours in salivary gl<strong>and</strong> cysts. Pathol Res Pract192:1211–1217174. Seifert G (1996) Classification <strong>and</strong> differential diagnosis <strong>of</strong>clear <strong>and</strong> basal cell tumors <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. Semin DiagnPathol 13:95–103175. Seifert G (1997) Primäre Speicheldrüsentumoren in Lymphknotender Parotis: Bericht über 3 Fälle und Übersicht überdie Literatur. Pathologe 18:141–146176. Seifert G, Miehlke A, Haubrich J, Chilla R (1986) Diseases <strong>of</strong><strong>the</strong> salivary gl<strong>and</strong>s. Thieme, Stuttgart177. Seifert G, Thomsen S, Donath K (1981) Bilateral dysgeneticpolycystic parotid gl<strong>and</strong>s: morphological analysis <strong>and</strong> differentialdiagnosis <strong>of</strong> a rare disease <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. VirchowsArch A Pathol Anat Histol 390:273–288178. Servillo G, Della Fazia MA, Sassone-Corsi P (2002) CouplingcAMP signaling to transcription in <strong>the</strong> liver: pivotal role <strong>of</strong>CREB <strong>and</strong> CREM. Exp Cell Res 275:143–154179. Sheldon WH (1943) So-called mixed tumors <strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong>s. Arch Pathol 35:1–20180. Shintaku M, Honda T (1997) Identification <strong>of</strong> oncocytic lesions<strong>of</strong> salivary gl<strong>and</strong>s by anti-mitochondrial immunohistochemistry.Histopathology 31:408–411181. Simpson RHW (1994) Classification <strong>of</strong> tumours <strong>of</strong> salivarygl<strong>and</strong>s. Histopathology 24:187–191182. Simpson RHW (1999) Clear cell tumors. Rev Esp Patol32:432–433183. Simpson RHW (2002) Myoepi<strong>the</strong>lial tumours <strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong>s. Curr Diagn Pathol 8:328–337184. Simpson RHW (2004) Case 22, slide seminar head <strong>and</strong> neckpathology. II. Intercontinental Congress <strong>of</strong> <strong>Pathology</strong>, Iguassu,Brazil, 2004185. Simpson RHW, Clarke TJ, Sarsfield PTL, Babajews AV (1991)Salivary duct adenocarcinoma. Histopathology 18:229–235186. Simpson RHW, Clarke TJ, Sarsfield PTL, Gluckman PGC(1991) Epi<strong>the</strong>lial-myoepi<strong>the</strong>lial carcinoma <strong>of</strong> salivary gl<strong>and</strong>s.J Clin Pathol 44:419–423187. Simpson RHW, Clarke TJ, Sarsfield PTL, Gluckman PGC,Babajews AV (1991) Polymorphous low grade adenocarcinoma<strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s: a clinicopathological comparisonwith adenoid cystic carcinoma. Histopathology 19:121–129188. Simpson RHW, Di Palma S (2003) Myoepi<strong>the</strong>lial tumour <strong>of</strong>salivary gl<strong>and</strong>s. In: Gale N, Luzan B (eds) Proceedings in<strong>Head</strong> <strong>and</strong> <strong>Neck</strong> <strong>Pathology</strong>. Institute <strong>of</strong> <strong>Pathology</strong> Faculty <strong>of</strong>Medicine, University <strong>of</strong> Lubljana, Lubljana 4–6 September2003, pp 92–98189. Simpson RHW, Jones H, Beasley P (1995) Myoepi<strong>the</strong>lioma <strong>of</strong><strong>the</strong> salivary gl<strong>and</strong>s: a true entity? Histopathology 27:1–9190. Simpson RHW, Prasad A, Lewis JE, Skálová A, David L(2003) Mucin-rich variant <strong>of</strong> salivary duct carcinoma: a


Major <strong>and</strong> Minor Salivary Gl<strong>and</strong>s Chapter 5 169clinicopathological study <strong>of</strong> four cases. Am J Surg Pathol27:1070–1079191. Simpson RHW, Reis-Filho JS, Pereira EM, Ribeiro AC, AbdulkadirA (2002) Polymorphous low grade adenocarcinoma<strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s with transformation to high grade carcinoma.Histopathology 41:250–259192. Simpson RHW, Robertson NJ, Arora DS, Saunders MW(1996) Salivary duct carcinoma: a cytological <strong>and</strong> histopathologicalstudy. Cytopathology 7:141–144193. Simpson RHW, Sarsfield PTL, Clarke T, Babajews AV (1990)Clear cell carcinoma <strong>of</strong> minor salivary gl<strong>and</strong>s. Histopathology17:433–438194. Simpson RHW, Sarsfield PTL (1997) Benign <strong>and</strong> malignantlymphoid lesions <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. Curr Diagn Pathol4:91–99195. Simpson RHW, Skálová A (1997). Metastatic carcinoma <strong>of</strong><strong>the</strong> prostate presenting as a parotid tumour. Histopathology30:70–74196. Skálová A, Lehtonen H, von Boguslawsky K, Leivo I (1994)Prognostic significance <strong>of</strong> cell proliferation in mucoepidermoidcarcinoma <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>: clinicopathologicalstudy using MIB1 antibody in paraffin sections. Hum Pathol25:929–935197. Skálová A, Leivo I, Michal M, Saksela E (1992) Analysis <strong>of</strong>collagen isotopes in crystalloid structures <strong>of</strong> salivary gl<strong>and</strong>tumors. Hum Pathol 23:748–754198. Skálová A, Leivo I, von Boguslawsky K, Saksela E (1994) Cellproliferation correlates with prognosis in acinic cell carcinomas<strong>of</strong> salivary gl<strong>and</strong> origin. Immunohistochemical study <strong>of</strong>30 cases using <strong>the</strong> MIB1 antibody in formalin-fixed paraffinsections. J Pathol 173:13–21199. Skálová A, Leivo I, Wolf H, Fakan F (2000) Oncocytic cystoadenoma<strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong> with tyrosine-rich crystals.Pathol Res Pract 196:849–851200. Skálová A, Michal M, Simpson RHW, Stárek I, Přádná J,Pfaltz M (2002) Sclerosing polycystic adenosis <strong>of</strong> parotidgl<strong>and</strong> with dysplasia <strong>and</strong> ductal carcinoma in situ: report <strong>of</strong>three cases with immunohistochemical <strong>and</strong> ultrastructuralexamination. Virchows Arch 440:29–35201. Skálová A, Simpson RHW, Lehtonen H, Leivo I (1997) Assessment<strong>of</strong> proliferative activity using <strong>the</strong> MIB1 antibodyhelps to distinguish polymorphous low grade adenocarcinomafrom adenoid cystic carcinoma <strong>of</strong> salivary gl<strong>and</strong>s. PatholRes Pract 193:695–703202. Skálová A, Stárek I, Kučerová V, Szépe P, Plank L (2001) Salivaryduct carcinoma – a highly aggressive salivary gl<strong>and</strong> tumorwith HER-2/neu oncoprotein overexpression. PatholRes Pract 197:621–626203. Skálová A, Stárek I, Simpson RHW, Kučerová V, DvorackovaJ, Čuřik R, Duskova M (2001) Spindle cell myoepi<strong>the</strong>lial tumours<strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong> with extensive lipomatous metaplasia.Virchows Arch 439:762–767204. Skálová A, Starek I, Vanecek T, Kucerova V, Plank L, SzepeP, Di Palma S, Leivo I (2003) Expression <strong>of</strong> HER-2/neu gene<strong>and</strong> protein in salivary duct carcinomas <strong>of</strong> parotid gl<strong>and</strong> asrevealed by immun<strong>of</strong>luorescence in situ hybridization <strong>and</strong>immunohistochemistry. Histopathology 42:1–9205. Škorpil F (1940) Über das Speicheldrüsenadenom. VirchowsArch A Anat Pathol 306:714–736206. Slootweg PJ (1993) Low-grade adenocarcinoma <strong>of</strong> <strong>the</strong> oralcavity: polymorphous or papillary? J Oral Pathol Med22:327–330207. Smyth AG, Ward-Booth RP, High AS (1993) Polycystic disease<strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>s: two familial cases. Br J Oral Maxill<strong>of</strong>acSurg 31:38–40208. Soares J (1999) Progression to low-grade <strong>and</strong> high-grade salivarygl<strong>and</strong> tumors. Rev Esp Patol 32:434–435209. Spiro RH, Huvos AG, Strong EW (1977) Malignant mixed tumor<strong>of</strong> salivary origin: a clinicopathologic study <strong>of</strong> 146 cases.Cancer 39:388–396210. Spiro RH, Huvos AG (1992) Stage means more than grade inadenoid cystic carcinoma. Am J Surg Pathol 164:623–628211. Sredni ST, da Cunha IW, de Carvalho Filho NP, Magrin J,Pinto CA, Lopes LF (2004) Endodermal sinus tumour <strong>of</strong> <strong>the</strong>parotid gl<strong>and</strong> in a child. Pediatr Dev Pathol 7:77–80212. Stallmach I, Zenklusen P, Komminoth P, Schmid S, Perren A,Roos M, Jianming Z, Heitz PU, Pfaltz M (2002) Loss <strong>of</strong> heterozygosityat chromosome 6q23-25 correlates with clinical<strong>and</strong> histologic parameters in salivary gl<strong>and</strong> adenoid cysticcarcinoma. Virchows Arch 440:77–84213. Stanley RJ, Weil<strong>and</strong> LH, Pearson BW (1988) Dedifferentiatedacinic cell (acinous) carcinoma <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 98:155–161214. Stephen J, Batsakis JG, Luna MA, von der Heyden U, Byers RM(1986) True malignant mixed tumors (carcinosarcoma) <strong>of</strong> salivarygl<strong>and</strong>s. Oral Surg Oral Med Oral Pathol 61:597–602215. Sternlicht MD, Barsky SH (1997) The myoepi<strong>the</strong>lial defense:a host defense against cancer. Med Hypo<strong>the</strong>ses 48:37–46216. Su AI, Welsh JB, Sapinoso LM, Kern SG, Dimitrov P, LappH, Schultz PG, Powell SM, Moskaluk CA, Frierson HF Jr,Hampton GM (2001) Molecular classification <strong>of</strong> human carcinomasby use <strong>of</strong> gene expression signatures. Cancer Res61:7388–7393217. Suchy BH, Wolf SR (2000) Bilateral mucosa-associated lymphoidtissue lymphoma <strong>of</strong> <strong>the</strong> parotid gl<strong>and</strong>. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 126:224–226218. Suzuki H, Inoue K, Fujioka Y, Ishikura H, Furuta Y, Fukuda S(1998) Myoepi<strong>the</strong>lial carcinoma with predominance <strong>of</strong> plasmacytoidcells arising in a pleomorphic adenoma <strong>of</strong> <strong>the</strong> parotidgl<strong>and</strong>. Histopathology 32:86–87219. Takata T, Ogawa I, Nikai H (1989) Sebaceous carcinoma <strong>of</strong><strong>the</strong> parotid gl<strong>and</strong>: an immunohistochemical <strong>and</strong> ultrastructuralstudy. Virchows Arch A Pathol Anat 414:459–464220. Takahashi H, Fujita S, Okabe H, Tsuda N, Tezuka F (1991)Immunohistochemical characterization <strong>of</strong> basal cell adenomas<strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>. Pathol Res Pract 187:145–156221. Takahashi H, Fujita S, Okabe H (1992) Distribution <strong>of</strong> tissuemarkers in acinic cell carcinomas <strong>of</strong> salivary gl<strong>and</strong>. PatholRes Pract 188:692–700222. Therkildsen MH, Christensen N, Andersen LJ, Larsen S,Katholm M (1992) Malignant Warthin’s tumour: a casestudy. Histopathology 21:167–171223. Tortoledo ME, Luna MA, Batsakis JG (1984) Carcinomas expleomorphic adenoma <strong>and</strong> malignant mixed tumors: histomorphologicindexes. Arch Otolaryngol 110:172–186224. Tse LL, Finkelstein SD, Siegler RW, Barnes L (2004) Osteoclast-typegiant cell neoplasm <strong>of</strong> salivary gl<strong>and</strong>. A microdissection-basedcomparative genotyping assay <strong>and</strong> literaturereview. Am J Surg Pathol 28:953–961225. Vargas H, Sudilovsky D, Kaplan MJ, Regezi JA, Weidner N(1997) Mixed tumor, polymorphous low-grade adenocarcinoma<strong>and</strong> adenoid cystic carcinoma <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>:pathogenic implications <strong>and</strong> differential diagnosis by Ki-67(MIB1), BCL2, <strong>and</strong> S-100 immunohistochemistry. Appl Immunohistochem5:8–16226. Vessecchia G, Di Palma S, Giardini R (1995) Subm<strong>and</strong>ibulargl<strong>and</strong> metastasis <strong>of</strong> breast carcinoma: a case report <strong>and</strong> review<strong>of</strong> <strong>the</strong> literature. Virchows Arch 427:349–351227. Viva E, Zorzi F, Annibale G, Stefini S, Baronchelli C, BonettiMF (1992) Endodermal sinus (yolk sac) tumour <strong>of</strong> <strong>the</strong> parotidgl<strong>and</strong>, a case report. Int J Pediatr Otorhinolaryngol24:269–274228. Waldron CA (1991) Mixed tumor (pleomorphic adenoma)<strong>and</strong> myoepi<strong>the</strong>lioma. In: Ellis GL, Auclair PL, Gnepp DR(eds) Surgical pathology <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. Saunders,Philadelphia, pp 165–186229. Waldron CA, el M<strong>of</strong>ty SK, Gnepp DR (1988) Tumors <strong>of</strong> <strong>the</strong>intraoral minor salivary gl<strong>and</strong>s: a demographic <strong>and</strong> histologicstudy <strong>of</strong> 426 cases. Oral Surg Oral Med Oral Pathol66:323–333230. Walt JD van der, Leake J (1987) Granulomatous sialadenitis<strong>of</strong> <strong>the</strong> major salivary gl<strong>and</strong>s. A clinicopathological study <strong>of</strong>57 cases. Histopathology 11:131–144231. Wanamaker JR, Kraus DH, Biscotti CV, Eliachar I (1994) Undifferentiatednasopharyngeal carcinoma presenting as a parotidmass. <strong>Head</strong> <strong>Neck</strong> 16:589–593


170 S. Di Palma · R.H.W. Simpson · A. Skalova · I. Leivo232. Wenig BM, Hitchcock CL, Ellis GL, Gnepp DR (1992) Metastasizingmixed tumor <strong>of</strong> salivary gl<strong>and</strong>s: a clinicopathologic <strong>and</strong>flow cytometric analysis. Am J Surg Pathol 16:845–858233. Werning JT (1991) Infectious <strong>and</strong> systemic diseases. In: EllisGL, Auclair PL, Gnepp DR (eds) Surgical pathology <strong>of</strong> <strong>the</strong>salivary gl<strong>and</strong>s. Saunders, Philadelphia, pp 39–59234. Wilson RW, Moran CA (1997) Epi<strong>the</strong>lial-myoepi<strong>the</strong>lial carcinoma<strong>of</strong> <strong>the</strong> lung: immunohistochemical <strong>and</strong> ultrastructuralobservations <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Hum Pathol28:631–635235. Worner GC, Reis PP, Makitie AA, Sukhai MA, Arora S, JurisicaI, Wells RA, Gullane P, Irish J, Kamel-Reid S (2004)Current applications <strong>of</strong> microarrays in head <strong>and</strong> neck cancerresearch. Laryngoscope 114:241–248236. Zarbo RJ, Prasad AR, Regezi JA, Gown AM, Savera AT (2000)Salivary gl<strong>and</strong> basal cell <strong>and</strong> canalicular adenomas: immunohistochemicaldemonstration <strong>of</strong> myoepi<strong>the</strong>lial cell participation<strong>and</strong> morphogenetic considerations. Arch Pathol124:401–405237. Zarbo RJ, Regezi JA, Batsakis JG (1986) S-100 protein in salivarygl<strong>and</strong> tumors: an immunohistochemical study <strong>of</strong> 129cases. <strong>Head</strong> <strong>Neck</strong> Surg 8:268–2755


Chapter 6Nasopharynx<strong>and</strong> Waldeyer’s RingS. Regauer6Contents6.1 Embryological Development<strong>of</strong> <strong>the</strong> Nasopharynx <strong>and</strong> Waldeyer’s Ring . . . . . . 1726.2 Nasopharynx . . . . . . . . . . . . . . . . . . . . . . . 1736.2.1 Anatomy <strong>and</strong> Histology . . . . . . . . . . . . . . . . 1736.2.2 Congenital Developmental Anomalies . . . . . . . . 1736.2.2.1 Nasopharyngeal Branchial Cleft Cysts . . . . . . . . 1736.2.2.2 Tornwaldt’s Cyst . . . . . . . . . . . . . . . . . . . . . 1736.2.2.3 Rathke’s Cleft Cyst/Ectopic Pituitary Tissue . . . . 1746.2.2.4 Craniopharyngioma . . . . . . . . . . . . . . . . . . 1746.2.2.5 Heterotopic Brain Tissue/Encephalocele . . . . . . 1746.2.3 Congenital Tumours . . . . . . . . . . . . . . . . . . 1746.2.3.1 Salivary Gl<strong>and</strong> Anlage Tumour . . . . . . . . . . . . 1756.2.3.2 Hairy Polyp . . . . . . . . . . . . . . . . . . . . . . . 1756.2.3.3 Congenital Nasopharyngeal Teratoma . . . . . . . . 1756.2.4 Benign Tumours <strong>and</strong> Tumour-Like Lesions . . . . . 1756.2.4.1 Nasopharyngeal Angi<strong>of</strong>ibroma . . . . . . . . . . . . 1756.2.4.2 Respiratory Epi<strong>the</strong>lialAdenomatoid Hamartoma . . . . . . . . . . . . . . . 1786.2.4.3 Nasopharyngeal Inverted Papilloma . . . . . . . . . 1786.2.4.4 Solitary Fibrous Tumour . . . . . . . . . . . . . . . . 1796.2.4.5 Paraganglioma . . . . . . . . . . . . . . . . . . . . . . 1796.2.4.6 Meningioma . . . . . . . . . . . . . . . . . . . . . . . 1796.2.4.7 Gl<strong>and</strong>ular Retention Cysts . . . . . . . . . . . . . . . 1796.2.5 Nasopharyngeal Carcinoma . . . . . . . . . . . . . . 1806.2.5.1 Non-Keratinising NasopharyngealCarcinoma . . . . . . . . . . . . . . . . . . . . . . . . 1806.2.5.2 Keratinising Nasopharyngeal Carcinoma . . . . . . 1826.2.6 Nasopharyngeal Adenocarcinoma . . . . . . . . . . 1826.2.6.1 Salivary Gl<strong>and</strong>-Type Adenocarcinoma<strong>of</strong> <strong>the</strong> Nasopharynx . . . . . . . . . . . . . . . . . . . 1826.2.6.2 Papillary Adenocarcinoma<strong>of</strong> <strong>the</strong> Nasopharynx . . . . . . . . . . . . . . . . . . . 1826.2.7 Malignant Non-Epi<strong>the</strong>lial Tumours<strong>of</strong> <strong>the</strong> Nasopharynx . . . . . . . . . . . . . . . . . . . 1836.2.7.1 Chordoma . . . . . . . . . . . . . . . . . . . . . . . . 1836.2.7.2 Sarcoma . . . . . . . . . . . . . . . . . . . . . . . . . . 1836.3 Waldeyer’s Ring . . . . . . . . . . . . . . . . . . . . . 1836.3.1 Anatomy <strong>and</strong> Histology <strong>of</strong> Waldeyer’s Ring . . . . . 1836.3.2 Congenital Anomalies <strong>of</strong> Waldeyer’s Ring . . . . . . 1846.3.3 Tonsillitis . . . . . . . . . . . . . . . . . . . . . . . . . 1846.3.3.1 Bacterial Tonsillitis . . . . . . . . . . . . . . . . . . . 1846.3.3.2 Viral Tonsillitis . . . . . . . . . . . . . . . . . . . . . 1856.3.4 Benign Tumours <strong>of</strong> Waldeyer’s Ring . . . . . . . . . 1876.3.4.1 Squamous Papilloma . . . . . . . . . . . . . . . . . . 1876.3.4.2 Lymphangiomatous Tonsillar Polyp . . . . . . . . . 1876.3.5 Carcinomas <strong>of</strong> Waldeyer’s Ring . . . . . . . . . . . . 1876.3.6 Malignant Lymphomas <strong>of</strong> Waldeyer’s Ring . . . . . 1896.3.6.1 Mantle Cell Lymphoma . . . . . . . . . . . . . . . . 1896.3.6.2 Extranodal Marginal Zone B-Cell Lymphoma<strong>of</strong> Mucosa-Associated Lymphoid Tissue . . . . . . . 1906.3.6.3 Extranodal NK/T-Cell Lymphoma,Nasal Type . . . . . . . . . . . . . . . . . . . . . . . . 1906.3.6.4 Hodgkin’s Lymphoma . . . . . . . . . . . . . . . . . 1906.3.6.5 Extramedullary Plasmacytoma . . . . . . . . . . . . 1906.3.7 Systemic Disease Affecting Waldeyer’s Ring . . . . 190References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191


172 S. Regauer66.1 Embryological Development<strong>of</strong> <strong>the</strong> Nasopharynx<strong>and</strong> Waldeyer’s RingThe pharynx is divided into an upper portion, <strong>the</strong> nasopharynx,<strong>and</strong> a lower portion consisting <strong>of</strong> <strong>the</strong> oropharynx<strong>and</strong> hypopharynx. The nasopharynx is locatedabove <strong>the</strong> s<strong>of</strong>t palate <strong>and</strong> communicates with <strong>the</strong> nasalpassages. The oropharynx extends from <strong>the</strong> s<strong>of</strong>t palate<strong>and</strong> <strong>the</strong> velum palatinum to <strong>the</strong> epiglottis. The hypopharynxextends from <strong>the</strong> tip <strong>of</strong> <strong>the</strong> epiglottis to <strong>the</strong>lower margin <strong>of</strong> <strong>the</strong> cricoid cartilage. The term Waldeyer’sring refers to <strong>the</strong> ring <strong>of</strong> lymphoid tissues extendingthroughout <strong>the</strong> naso- <strong>and</strong> oropharynx <strong>and</strong> includes <strong>the</strong>palatine, pharyngeal, lingual <strong>and</strong> tubal tonsils.The earliest embryological stage relevant for <strong>the</strong> development<strong>of</strong> <strong>the</strong> nasopharynx is around <strong>the</strong> 3rd week <strong>of</strong>gestation. The embryo has already developed all threegerm layers <strong>and</strong> consists <strong>of</strong> <strong>the</strong> notochord with a notochordallumen [134]. At <strong>the</strong> cranial end, <strong>the</strong> notochordis still fused with <strong>the</strong> ecto- <strong>and</strong> endodermal germ layers,which form <strong>the</strong> bilaminar oropharyngeal (or buccopharyngeal)membrane. The first process in <strong>the</strong> formation<strong>of</strong> <strong>the</strong> pharynx is <strong>the</strong> development <strong>of</strong> a primitive mouth(or stomodeum) <strong>and</strong> a primitive pharynx during <strong>the</strong> 4thweek via rupture <strong>of</strong> <strong>the</strong> oropharyngeal membrane. Atthat time, <strong>the</strong> primitive nasal cavities <strong>and</strong> <strong>the</strong> nasopharynxare still separated from <strong>the</strong> oral cavity by <strong>the</strong> primarypalate <strong>and</strong> <strong>the</strong> oronasal membrane. The oronasalmembrane ruptures around week 6, bringing <strong>the</strong> nasal<strong>and</strong> oral cavity into continuity <strong>and</strong> forming <strong>the</strong> primitivechoanae. During <strong>the</strong> posterior extension <strong>of</strong> <strong>the</strong> primarypalate <strong>and</strong> development <strong>of</strong> <strong>the</strong> secondary palate,<strong>the</strong> choanae become located posteriorly <strong>and</strong> connect <strong>the</strong>newly formed nasal cavities <strong>and</strong> nasopharynx. The development<strong>of</strong> <strong>the</strong> pituitary gl<strong>and</strong> (or hypophysis) beginsaround <strong>the</strong> middle <strong>of</strong> <strong>the</strong> 4th week with an upward proliferation<strong>of</strong> <strong>the</strong> ectodermal ro<strong>of</strong> <strong>of</strong> <strong>the</strong> stomodeum, <strong>the</strong>Rathke’s pouch (or hypophyseal duct), <strong>and</strong> a downgrowthfrom <strong>the</strong> diencephalon called <strong>the</strong> neurohypophyseal bud.The Rathke’s pouch passes through <strong>the</strong> chondrificationcentres <strong>of</strong> <strong>the</strong> developing sphenoid bones. By <strong>the</strong> 5thweek, both portions <strong>of</strong> <strong>the</strong> pituitary gl<strong>and</strong> have come incontact <strong>and</strong> <strong>the</strong> Rathke’s pouch becomes constricted atits attachment to <strong>the</strong> oronasal epi<strong>the</strong>lium. The stalk degenerates<strong>and</strong> <strong>the</strong> Rathke’s pouch involutes to a series <strong>of</strong>microcysts, which persist throughout adult life in <strong>the</strong> pituitarygl<strong>and</strong>. Occasionally, <strong>the</strong>y can be recognised macroscopicallyby a zone <strong>of</strong> colloid cysts. Symptomatic enlargementleads to <strong>the</strong> formation <strong>of</strong> a Rathke’s cleft cyst(see Sect. 6.2.2.3). Around <strong>the</strong> same time, during <strong>the</strong> involution<strong>of</strong> <strong>the</strong> notochord, a pharyngeal bursa is formedtemporarily at <strong>the</strong> site <strong>of</strong> early communication between<strong>the</strong> notochord <strong>and</strong> <strong>the</strong> ro<strong>of</strong> <strong>of</strong> <strong>the</strong> pharynx. This ectodermallyderived pharyngeal bursa normally obliteratesduring <strong>the</strong> 6th gestational week. A persistent pharyngealbursa in adults is located at <strong>the</strong> posterior median wall<strong>of</strong> <strong>the</strong> nasopharynx above <strong>the</strong> superior pharyngeal constrictormuscles at <strong>the</strong> lower end <strong>of</strong> <strong>the</strong> pharyngeal tonsil<strong>and</strong> is known as Tornwaldt’s cyst (see Sect. 6.2.2.2).Remnants <strong>of</strong> <strong>the</strong> notochord give rise to cranial chordomas(see Sect. 6.2.7.1).The oropharynx, mouth <strong>and</strong> larynx develop from<strong>the</strong> pharyngeal (or branchial) apparatus during <strong>the</strong>4th <strong>and</strong> 5th weeks <strong>of</strong> gestation. The growth <strong>of</strong> <strong>the</strong> forebrain<strong>and</strong> <strong>the</strong> development <strong>of</strong> <strong>the</strong> pharyngeal/branchialapparatus produce a prominent elevation <strong>of</strong> <strong>the</strong>head with a quite distinct first <strong>and</strong> second pharyngealarch around day 24. At <strong>the</strong> end <strong>of</strong> <strong>the</strong> 4th embryonicweek, four well-defined <strong>and</strong> two rudimentary bilateralpairs <strong>of</strong> pharyngeal arches are separated by <strong>the</strong> pharyngealgrooves. Each arch consists <strong>of</strong> a core <strong>of</strong> embryonicmesenchyme with an artery, a cartilage rod, a nerve<strong>and</strong> a muscular component, <strong>and</strong> is covered externallyby ectoderm <strong>and</strong> internally by endoderm. The ectodermallyderived pharyngeal arches <strong>and</strong> grooves support<strong>the</strong> lateral walls <strong>of</strong> <strong>the</strong> primitive pharynx. The innerlining consists <strong>of</strong> endoderm with balloon-like diverticulicalled pharyngeal pouches, which are also presentin four well-defined pairs. The second pharyngealpouch is <strong>the</strong> major contributor to <strong>the</strong> formation <strong>of</strong><strong>the</strong> pharynx <strong>and</strong> is largely obliterated when <strong>the</strong> palatinetonsils develop around weeks 12–14 post-conception.A part <strong>of</strong> <strong>the</strong> cavity <strong>of</strong> <strong>the</strong> second pouch remainsas <strong>the</strong> intratonsillar cleft (or tonsillar fossa) in <strong>the</strong> palatinetonsils (see Sect. 6.3.1). The neural crest-derivedmesenchyme will form most <strong>of</strong> <strong>the</strong> skeletal (cartilage<strong>and</strong> bone) <strong>and</strong> connective tissue structure <strong>of</strong> <strong>the</strong> head<strong>and</strong> neck, but <strong>the</strong> original mesenchyme <strong>of</strong> <strong>the</strong> secondarch forms <strong>the</strong> blood vessels <strong>and</strong> skeletal musculature<strong>of</strong> <strong>the</strong> pharynx. The nerve supply <strong>of</strong> <strong>the</strong> pharynx developsfrom <strong>the</strong> 3rd pharyngeal arch (IX glossopharyngealnerve). The adult vascular pattern <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck depends on a complex transformation <strong>of</strong> <strong>the</strong> pharyngealapparatus with involution <strong>and</strong> obliteration <strong>of</strong><strong>the</strong> early vessels. Incomplete involution, particularly<strong>of</strong> <strong>the</strong> first pharyngeal artery, has been postulated tobe responsible for <strong>the</strong> development <strong>of</strong> nasopharyngealangi<strong>of</strong>ibromas (see Sect. 6.2.4.1). The auditory (Eustachian)tube is derived from <strong>the</strong> first pharyngeal arch<strong>and</strong> pouch. Incomplete regression may be responsiblefor <strong>the</strong> occurrence <strong>of</strong> hairy polyps (see Sect. 6.2.3.2).The tongue also begins to develop at <strong>the</strong> end <strong>of</strong> <strong>the</strong> 4thweek. The oral part <strong>of</strong> <strong>the</strong> tongue is derived from <strong>the</strong>first pharyngeal arch; <strong>the</strong> posterior pharyngeal tonguedevelops by fusion <strong>of</strong> <strong>the</strong> ventromedial parts <strong>of</strong> <strong>the</strong> second<strong>and</strong> <strong>the</strong> third pharyngeal arch. The tissues derivedfrom <strong>the</strong> second pharyngeal arch are gradually overgrownby <strong>the</strong> third pharyngeal arch.The lymphoid tissues <strong>of</strong> Waldeyer’s ring develop between<strong>the</strong> 14th <strong>and</strong> 18th weeks <strong>of</strong> gestation. The development<strong>of</strong> <strong>the</strong> pharyngeal tonsil begins from an anlage


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 173consisting <strong>of</strong> longitudinal folds on <strong>the</strong> dorsal wall <strong>of</strong> <strong>the</strong>nasopharynx around <strong>the</strong> 12–14th gestational week. Thedevelopment <strong>of</strong> <strong>the</strong> palatine tonsils begins with a proliferation<strong>of</strong> <strong>the</strong> endodermal epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong> second pharyngealpouch down into <strong>the</strong> surrounding mesenchyme,forming <strong>the</strong> epi<strong>the</strong>lium-lined crypts. In <strong>the</strong> connectivetissue, mesenchymal cells <strong>of</strong> <strong>the</strong> second pharyngealpouch form so-called condensation centres. The firstprimary follicles can be localised around week 14. Theparafollicular areas develop into T-cell areas <strong>and</strong> precursors<strong>of</strong> interdigitating cells can be identified. Aroundweek 16, <strong>the</strong> epi<strong>the</strong>lium shows <strong>the</strong> first signs <strong>of</strong> cornification,<strong>and</strong> <strong>the</strong> lymphocytic infiltration <strong>of</strong> <strong>the</strong> epi<strong>the</strong>liumoccurs [59, 60].6.2 Nasopharynx6.2.1 Anatomy <strong>and</strong> HistologyThe nasopharynx is bordered anteriorly by <strong>the</strong> nasalchoanae <strong>and</strong> nasal cavities. The ro<strong>of</strong> <strong>and</strong> posterior wall<strong>of</strong> <strong>the</strong> nasopharynx form an arch just below <strong>the</strong> base <strong>of</strong><strong>the</strong> skull with <strong>the</strong> sphenoid sinus <strong>and</strong> <strong>the</strong> floor <strong>of</strong> <strong>the</strong>sella turcica. The postero-superior ro<strong>of</strong> contains <strong>the</strong>(naso)pharyngeal tonsil. At its lower end in <strong>the</strong> posteriormidline lies a blind recess known as <strong>the</strong> pharyngealbursa. The lateral walls <strong>of</strong> <strong>the</strong> nasopharynx contain <strong>the</strong>pharyngeal ostia <strong>of</strong> <strong>the</strong> auditory tubes, which are surroundedby small aggregates <strong>of</strong> lymphoid tissue, <strong>the</strong>tubal tonsil. The cartilage <strong>of</strong> <strong>the</strong> auditory tube protrudesabove <strong>the</strong> ostium <strong>and</strong> is called <strong>the</strong> torus tubarius, behindwhich <strong>the</strong> Rosenmüller fossa is located. The floor <strong>of</strong><strong>the</strong> nasopharynx is formed by <strong>the</strong> s<strong>of</strong>t palate <strong>and</strong> uvula.The anterior <strong>and</strong> cranial portions <strong>of</strong> <strong>the</strong> nasopharynxare lined by respiratory mucosa with ciliated columnarepi<strong>the</strong>lium with goblet cells <strong>and</strong> foci <strong>of</strong> metaplasticsquamous epi<strong>the</strong>lium. Squamous mucosa predominatesin <strong>the</strong> lower nasopharynx adjacent to <strong>the</strong> oropharynx.Small seromucinous gl<strong>and</strong>s <strong>and</strong> aggregates <strong>of</strong> lymphoidtissue are present in <strong>the</strong> submucosa throughout <strong>the</strong> nasopharynxas a normal finding without qualifying as“chronic inflammation”.6.2.2 CongenitalDevelopmental AnomaliesCongenital developmental anomalies <strong>of</strong> <strong>the</strong> nasopharynxare rare <strong>and</strong> include choanal atresias <strong>and</strong> pharyngealstenosis. Bilateral occlusion has been reported inup to 40% <strong>of</strong> patients with choanal atresias. Bilateralatresias are typically osseous, unilateral atresias aremembranous. Pharyngeal stenosis is a paediatric disorderassociated with sleep apnoea <strong>and</strong> o<strong>the</strong>r crani<strong>of</strong>acialanomalies [108, 137]. Far more common are congenitalcysts <strong>and</strong> tumours arising from remnants <strong>of</strong> embryonictissue.6.2.2.1 NasopharyngealBranchial Cleft CystsNasopharyngeal branchial cleft cysts <strong>and</strong> fistulas arelocated in <strong>the</strong> lateral nasopharyngeal wall <strong>and</strong> can extendto <strong>the</strong> base <strong>of</strong> <strong>the</strong> skull through <strong>the</strong> parapharyngealspace. They occur unilaterally or bilaterally <strong>and</strong> have apostulated second pharyngeal arch origin. Most nasopharyngealbranchial cleft cysts are clinically silent duringchildhood <strong>and</strong> discovery may be delayed into earlyadulthood, when chronic inflammation induces proliferation<strong>of</strong> <strong>the</strong> lymphoid tissues. This produces a massresulting in respiratory difficulties <strong>and</strong> nasal obstruction.The cysts are lined with respiratory epi<strong>the</strong>lium.Chronic irritation <strong>and</strong> inflammation induces squamousmetaplasia. Surgical specimens <strong>of</strong> nasopharyngeal cystsremoved for symptomatic disease may <strong>the</strong>refore showexclusively squamous epi<strong>the</strong>lium <strong>and</strong> lymphoid hyperplasiain a fibrosed cyst wall [143, 177, 185].6.2.2.2 Tornwaldt’s CystA Tornwaldt’s cyst (or pharyngeal bursitis; named afterGustav Ludwig Tornwaldt, 1843–1910) is a dilatation <strong>of</strong> apersistent pharyngeal bursa in <strong>the</strong> posterior median wall<strong>of</strong> <strong>the</strong> nasopharynx above <strong>the</strong> superior pharyngeal constrictormuscles <strong>and</strong> at <strong>the</strong> lower end <strong>of</strong> <strong>the</strong> pharyngealtonsil caudally <strong>and</strong> posteriorly to where Rathke’s cleftcysts arise [128]. The cysts arise at <strong>the</strong> site <strong>of</strong> embryoniccommunication between <strong>the</strong> notochord <strong>and</strong> <strong>the</strong> ro<strong>of</strong> <strong>of</strong><strong>the</strong> pharynx <strong>and</strong> can be detected in up to 7% <strong>of</strong> adultsat routine autopsies [86]. Tornwaldt’s cysts are typicallyless than 1 cm in size <strong>and</strong> asymptomatic [92]. Obstruction<strong>of</strong> <strong>the</strong> bursal orifice results in a cystic dilatation.The cysts may become infected <strong>and</strong> inflamed with subsequentabscess formation. Symptomatic disease, termedTornwaldt’s disease, may present with nasal obstruction<strong>and</strong> nasopharyngeal drainage, dull occipital headaches,pain in <strong>the</strong> ears <strong>and</strong> neck muscles <strong>and</strong> occasionally neckmuscle stiffness [130]. Tornwaldt’s cysts are lined withtall, columnar, ciliated respiratory epi<strong>the</strong>lium, but inflammation<strong>and</strong> infection induces squamous metaplasia<strong>and</strong> fibrosis <strong>of</strong> <strong>the</strong> walls. Tornwaldt’s cyst may containvarying amounts <strong>of</strong> lymphoid tissue (Fig. 6.1). Most surgicalspecimens <strong>of</strong> symptomatic Tornwaldt’s cysts areei<strong>the</strong>r devoid <strong>of</strong> an epi<strong>the</strong>lial lining or lined with squamousepi<strong>the</strong>lium.


174 S. Regauer6Fig. 6.1. Tornwaldt’s cyst. Tornwaldt’s cysts are thin-walled cysts<strong>of</strong> varying sizes located in <strong>the</strong> nasopharyngeal submucosa. Thenasopharyngeal surface is covered ei<strong>the</strong>r by squamous epi<strong>the</strong>liumor respiratory epi<strong>the</strong>lium. The cysts are lined by respiratorytype epi<strong>the</strong>lium with varying extent <strong>of</strong> squamous metaplasia. Thecyst wall contains varying amounts <strong>of</strong> inflammatory cells <strong>and</strong> maybe fibrosed6.2.2.3 Rathke’s Cleft Cyst/Ectopic Pituitary TissueRathke’s cleft cysts (or Rathke’s cyst; named after MartinHeinrich Rathke, 1793–1860) arise from <strong>the</strong> Rathke’spouch (or hypophyseal duct), an ectodermal remnant <strong>of</strong><strong>the</strong> stomodeum that gives rise to <strong>the</strong> anterior lobe <strong>of</strong> <strong>the</strong>pituitary gl<strong>and</strong>. The Rathke’s pouch is normally obliteratedto become microscopic clefts, but remnants <strong>of</strong> <strong>the</strong>Rathke’s pouch are incidental findings in up to 33% <strong>of</strong> unselectedautopsies. Rathke’s cleft cysts can have diametersranging from 0.3 to 4 cm [90, 192]. The thin-walled cystsare lined with respiratory epi<strong>the</strong>lium with interspersedgoblet cells <strong>and</strong> are filled with a clear, colloid, mucinous,viscous, turbid or haemorrhagic fluid. In cases <strong>of</strong> inflammation,metaplastic non-keratinising squamous epi<strong>the</strong>lium<strong>and</strong> a mural lymphocytic infiltrate dominate [72, 90].Rupture induces a granulomatous <strong>and</strong> xanthomatous inflammationwith fibrosis, cholesterol needles <strong>and</strong> foreignbody cell reaction, <strong>and</strong> even amyloid deposition. Rathke’scleft cysts are virtually never removed intact <strong>and</strong> surgicalspecimens typically consist <strong>of</strong> inflamed fibrous tissue.The epi<strong>the</strong>lium is <strong>of</strong>ten elusive. Rare examples <strong>of</strong> Rathke’scleft cysts contain mural nodules <strong>of</strong> anterior pituitary tissueor concomitant pituitary adenoma. Most nasopharyngealpituitary adenomas represent nasopharyngealextension <strong>of</strong> intracranial pituitary adenomas. Primarynasopharyngeal pituitary adenomas are extremely rare<strong>and</strong> mostly non-functioning [110, 123]. Symptoms dependon <strong>the</strong> type <strong>of</strong> secreted hormone <strong>and</strong> <strong>the</strong> location <strong>of</strong><strong>the</strong> adenoma. Histologically, ectopic pituitary gl<strong>and</strong> tissuecan display <strong>the</strong> entire spectrum with chromophobe,acidophilic <strong>and</strong> basophilic cells [29].6.2.2.4 CraniopharyngiomaICD-O:9350/1All reported extracranial craniopharyngiomas (orRathke’s pouch tumour, craniopharyngeal duct tumour)were located in <strong>the</strong> nasopharynx, sella turcica <strong>and</strong> sphenoidsinus, <strong>and</strong> presented during <strong>the</strong> first two decades <strong>of</strong>life with headaches, nasal obstruction <strong>and</strong> epistaxis (forreviews see [18, 67]). Intracranial craniopharyngiomasoccur classically in an adamantinomatous <strong>and</strong> papillarysubtype. The majority <strong>of</strong> <strong>the</strong> extracranial nasopharyngealcraniopharyngiomas are <strong>of</strong> <strong>the</strong> adamantinomatoussubtype with cords <strong>of</strong> basaloid squamous cells <strong>and</strong> foci<strong>of</strong> squamous differentiation <strong>and</strong> horn pearls embeddedin fibrous stroma. O<strong>the</strong>r typical secondary changes arecysts filled with brown fluid, areas <strong>of</strong> necrosis, calcifications<strong>and</strong> cholesterol crystals. Treatment is surgical <strong>and</strong>supplemented with radio<strong>the</strong>rapy in <strong>the</strong> case <strong>of</strong> incompletelyresected tumours <strong>and</strong> recurrences.6.2.2.5 Heterotopic Brain Tissue/EncephaloceleEncephaloceles are herniations <strong>of</strong> brain through a congenitalopening <strong>of</strong> <strong>the</strong> skull. Glial heterotopias with <strong>and</strong>without encephaloceles cause neonatal airway obstruction<strong>and</strong> rhinorrhoea. Heterotopic brain in a nasopharyngeallocation is rare [168]. From a pathologist’s perspective,encephaloceles <strong>and</strong> brain heterotopias can bedistinguished only after correlation with <strong>the</strong> patient’sclinical <strong>and</strong> radiological findings. Histologically, matureneuroglial tissues are embedded in fibrovascularstroma. Encephaloceles/glial heterotopias involving <strong>the</strong>nasopharynx differ from <strong>the</strong> more common nasal gliomasin that <strong>the</strong>y contain ependyma, choroid plexus, retinalcomponents <strong>and</strong> occasionally neoplastic tissue, suchas an oligodendroglioma [14].6.2.3 Congenital TumoursAmong <strong>the</strong> tumourous developmental abnormalities arehamartomas, choristomas <strong>and</strong> teratomas. A hamartomais a benign tumour-like nodule composed <strong>of</strong> maturecells <strong>and</strong> tissues that normally occur in <strong>the</strong> affected part,but <strong>of</strong>ten with one element predominating. Choristomasrepresent a mass <strong>of</strong> histologically normal mature tissueslocated at a site where <strong>the</strong>se tissues do not normally occur.These tumours are also called aberrant rests or heterotopictissue. Teratomas are true neoplastic proliferationsmade up <strong>of</strong> mature or immature tissues in r<strong>and</strong>omorganisation derived from all three germ layers.


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 1756.2.3.1 Salivary Gl<strong>and</strong> Anlage TumourSalivary gl<strong>and</strong> anlage tumours are rare tumours <strong>of</strong> neonatesthat arise from minor salivary gl<strong>and</strong>s <strong>of</strong> <strong>the</strong> nasopharynx<strong>and</strong> are also known as congenital pleomorphicadenoma [13, 38, 73]. The firm, polypoid, pedunculatedmidline masses can reach up to 3 cm in size <strong>and</strong> causerespiratory distress <strong>and</strong> feeding problems. Salivarygl<strong>and</strong> anlage tumours are multinodular <strong>and</strong> usuallysolid tumours. Histologically, <strong>the</strong>y are characterised byan epi<strong>the</strong>lial proliferation imitating embryonic salivarygl<strong>and</strong>s (Fig. 6.2). The epi<strong>the</strong>lial proliferation can be extensivewith solid squamous areas with focal keratinisation,keratinised nests, cyst <strong>and</strong> pearls. Calcificationwithin <strong>the</strong> cysts occurs. Salivary gl<strong>and</strong> anlage tumoursmay contain branching ductal <strong>and</strong> gl<strong>and</strong>ular structures,occasionally with complex intraluminal papillations.The surrounding stroma may be loose <strong>and</strong> myxoid withnumerous inflammatory cells, but may also show somefibrosis. O<strong>the</strong>r salivary gl<strong>and</strong> anlage tumours consistpredominantly <strong>of</strong> densely packed sheet <strong>and</strong> nodules<strong>of</strong> small fusiform spindle cells with occasional regularmitoses. Rare keratinised duct <strong>and</strong> cystic structures areseen in <strong>the</strong>se areas. Haemorrhage <strong>and</strong> focal necrosis canbe seen [125]. Immunohistochemically, all cells stainpositive for salivary gl<strong>and</strong> amylase. The spindle cells areimmunoreactive with antibodies to vimentin, cytokeratins,EMA <strong>and</strong> smooth muscle actin, but are negative forS-100 <strong>and</strong> GFAP. The epi<strong>the</strong>lial structures stain for cytokeratin<strong>and</strong> EMA. The exact classification <strong>of</strong> <strong>the</strong> salivarygl<strong>and</strong> anlage tumour as a hamartoma or true (benign)neoplasm has not yet been resolved. Dehner <strong>and</strong>colleagues favour a hamartomatous origin, although <strong>the</strong>present name indicates a tumourous growth [38].6.2.3.2 Hairy PolypHairy polyps (or dermoids) are found in <strong>the</strong> naso- <strong>and</strong>oropharynx <strong>of</strong> neonates or young infants. About 60% <strong>of</strong><strong>the</strong> roughly 140 reported cases arose as single, pedunculatedor sessile, 0.5 to 6 cm polyps in <strong>the</strong> lateral vault<strong>of</strong> <strong>the</strong> nasopharynx near <strong>the</strong> Eustachian tube orifice<strong>and</strong> very rarely within <strong>the</strong> Eustachian tube <strong>and</strong> middleear [17, 78, 109]. The remaining cases occurred in <strong>the</strong>tonsillar region (for tonsillar <strong>and</strong> bilateral location seeSect. 6.3.2). Hairy polyps cause respiratory distress orfeeding problems. Simultaneous congenital abnormalitiessuch as cleft palate are more common than ipsilateralbranchial sinus, congenital atresia <strong>of</strong> <strong>the</strong> carotidartery, osteopetrosis <strong>and</strong> malformations <strong>of</strong> <strong>the</strong> auricle[36, 68]. Treatment is simple surgical resection. A hairypolyp is covered by keratinised or glycogenated squamousepi<strong>the</strong>lium containing hair with sebaceous gl<strong>and</strong>s<strong>and</strong> sweat gl<strong>and</strong>s. The mesenchymal core consists <strong>of</strong>mature fibroadipose tissue <strong>and</strong> blood vessels with occasionalsmooth muscle <strong>and</strong> striated muscle fibres. Thestalk may contain foci or plates <strong>of</strong> hyaline cartilage [27].Hairy polyps fulfil <strong>the</strong> definition <strong>of</strong> a choristoma, although<strong>the</strong> literature refers to hairy polyps mistakenly<strong>of</strong>ten as “teratoma”.6.2.3.3 CongenitalNasopharyngeal TeratomaICD-O:9080/1Congenital teratomas are most common in <strong>the</strong> saccrococcygealregion; less than one-third <strong>of</strong> all teratomasarise in <strong>the</strong> head <strong>and</strong> neck region. The nasopharynx isan exceptionally rare location [84, 167, 187]. Teratomasare ill-formed, lobular solid <strong>and</strong> cystic tumour massestypically diagnosed in <strong>the</strong> neonatal period with airwayobstruction. Mature teratomas are completely benign<strong>and</strong> consist <strong>of</strong> mature tissues <strong>of</strong> ecto-, meso- <strong>and</strong> endodermalderivation in a more or less organised fashion.Optimal treatment is complete excision. Immatureteratomas contain tissues <strong>of</strong> varying degrees <strong>of</strong> differentiation<strong>and</strong> maturation. Immature teratomas in infantshave an excellent prognosis, quite in contrast to adultpatients with immature teratomas.6.2.4 Benign Tumours<strong>and</strong> Tumour-Like Lesions6.2.4.1 Nasopharyngeal Angi<strong>of</strong>ibromaICD-O:9160/0Nasopharyngeal angi<strong>of</strong>ibromas (or juvenile nasopharyngealangi<strong>of</strong>ibromas) are rare benign tumours with anincidence <strong>of</strong> 0.5% <strong>of</strong> all head <strong>and</strong> neck tumours. Theyoccur exclusively in adolescent males <strong>and</strong> become clinicallyevident between 10 <strong>and</strong> 25 years <strong>of</strong> age with nosebleeds, respiratory distress, headaches or sometimesvisual disturbances. Nasopharyngeal angi<strong>of</strong>ibromasarise from <strong>the</strong> posterior lateral wall <strong>of</strong> <strong>the</strong> nasal cavitynear <strong>the</strong> pterygo-palatine fossa at <strong>the</strong> superior margin<strong>of</strong> <strong>the</strong> foramen sphenopalatinum <strong>and</strong> extend into adjacentstructures such as maxillary, sphenoid or ethmoidsinuses <strong>and</strong> <strong>the</strong> nasal cavity. The most advanced casesshow intracranial extension [153, 203]. The blood supply<strong>of</strong> nasopharyngeal angi<strong>of</strong>ibromas comes from <strong>the</strong> externalcarotid artery with <strong>the</strong> internal maxillary artery asfeeding branch. In a minority <strong>of</strong> cases, <strong>the</strong> feeding vesselsare <strong>the</strong> sphenopalatine artery <strong>and</strong> <strong>the</strong> ascendingpharyngeal artery [75, 153]. Diagnosis <strong>of</strong> a nasopharyngealangi<strong>of</strong>ibroma is based on clinical examination<strong>and</strong> computer tomography, which shows two consistentfeatures: 1) localisation in <strong>the</strong> posterior nasal cavity<strong>and</strong> pterygopalatine fossa, <strong>and</strong> 2) bone erosions <strong>of</strong> <strong>the</strong>


176 S. Regauer6abcdefg


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 177abcdefFig. 6.3. Nasopharyngeal angi<strong>of</strong>ibroma. a–b Nasopharyngeal angi<strong>of</strong>ibromasare large, white, firm, bulging tumours covered by respiratorymucosa. c–d Abnormal large muscular <strong>and</strong> smaller sinusoidalvessels are embedded in a fibrous <strong>and</strong> partly sclerotic stroma.The muscular vessels exhibit irregular muscle walls with varyingthickness <strong>and</strong> arbitrary arrangement <strong>of</strong> <strong>the</strong> individual smoothmuscle cells. e–f The stroma is ei<strong>the</strong>r fibrous or myxoid with stellateor spindled cellsFig. 6.2. Salivary gl<strong>and</strong> anlage tumour. a–c Salivary gl<strong>and</strong> anlagetumours have an intact surface epi<strong>the</strong>lium, which may be partlykeratinized. The branching epi<strong>the</strong>lial proliferations show ductsfilled with proteinaceous casts. The stroma is myxoid <strong>and</strong> containsnumerous plasma cells <strong>and</strong> lymphocytes. d–e There are solidsquamous areas with keratin-filled cysts <strong>and</strong> calcifications. f–gO<strong>the</strong>r areas are dominated by proliferations <strong>of</strong> spindle cells in solid,fascicular <strong>and</strong> nested arrangement with occasional ductal <strong>and</strong>cystic elements. Haemorrhage is common


178 S. Regauer6sphenopalatine foramen with extension to <strong>the</strong> uppermedial pterygoid plate [118]. Nasopharyngeal angi<strong>of</strong>ibromasare unencapsulated, lobulated, firm, grey totan tumours. Abnormal blood vessels <strong>of</strong> different sizes<strong>and</strong> irregular architecture in arbitrary arrangement areembedded in a my<strong>of</strong>ibroblastic stroma. The proportions<strong>of</strong> both components can vary considerably (Fig. 6.3).The vascular component can be divided into small sinusoidalvessels <strong>and</strong> large muscular vessels with irregular<strong>and</strong> incomplete smooth muscle layers with abrupttransitions from a muscular coat to an endo<strong>the</strong>lial celllining only. Irrespective <strong>of</strong> <strong>the</strong>ir architecture, all vesselsare lined with a continuous layer <strong>of</strong> single regular endo<strong>the</strong>lialcells. The stromal component varies in amount<strong>and</strong> cellularity. The majority <strong>of</strong> stromal fibroblasts areplump <strong>and</strong> stellate, o<strong>the</strong>rs assume an elongated spindledconfiguration. Mitoses are inconspicuous. The majority<strong>of</strong> stromal cells react with vimentin only, while a subpopulationis characterised by co-expression <strong>of</strong> vimentin<strong>and</strong> smooth muscle actin.Despite <strong>the</strong> body <strong>of</strong> literature, <strong>the</strong>re is still uncertaintyabout <strong>the</strong> aetiology <strong>of</strong> nasopharyngeal angi<strong>of</strong>ibromas.They have been called haemangioma <strong>and</strong> vascular hamartomaarising from ectopic vascular tissue <strong>of</strong> <strong>the</strong> inferiorturbinate. O<strong>the</strong>r <strong>the</strong>ories have included overgrowth<strong>of</strong> paraganglionic tissue, hyperplasia in response to allergicstimulus, fibromatosis, teratoma arising from <strong>the</strong>occipital plate, but also an <strong>and</strong>rogen-dependent neoplasticprocess due to an imbalance <strong>of</strong> <strong>the</strong> pituitary–<strong>and</strong>rogenitalsystem [9]. A concomitant presentation <strong>of</strong> nasopharyngealangi<strong>of</strong>ibromas <strong>and</strong> <strong>the</strong> familial adenomatouspolyposis syndrome has been reported in 4 out <strong>of</strong>825 patients at <strong>the</strong> Johns Hopkins’ Registry for familialcolonic polyposis [47, 62]. A mutation in <strong>the</strong> APC gene,however, was not demonstrated in 9 patients with nasopharyngealangi<strong>of</strong>ibroma, <strong>and</strong> a comparative genomichybridisation study describes a normal chromosome5 in 3 patients with nasopharyngeal angi<strong>of</strong>ibroma [70,170]. The same study reports gains on chromosome 8at <strong>the</strong> site <strong>of</strong> <strong>the</strong> genes for TGF-ß inducible early growthresponse <strong>and</strong> LYN (v-yes-1 Yamaguchi sarcoma viral-relatedoncogene homologue), <strong>and</strong> on chromosome 6, onwhich <strong>the</strong> gene for <strong>the</strong> vascular endo<strong>the</strong>lial growth factoris located [170]. The additional complex gains on <strong>the</strong>X chromosome <strong>and</strong> losses on <strong>the</strong> Y chromosome may explain<strong>the</strong> exclusive occurrence in male patients. The interpretation<strong>of</strong> nasopharyngeal angi<strong>of</strong>ibromas as a vascularmalformation has proved to be <strong>the</strong> most consistent<strong>the</strong>ory over <strong>the</strong> past few decades. A recent publicationproposes that nasopharyngeal angi<strong>of</strong>ibromas arise froman embryological vascular remnant <strong>of</strong> <strong>the</strong> first pharyngealarch artery, which normally regresses to a vascularplexus, giving rise to <strong>the</strong> maxillary artery [75, 153, 171].Incomplete involution leaves behind vascular remnantsin <strong>the</strong> lateral nasal wall in <strong>the</strong> area <strong>of</strong> <strong>the</strong> sphenopalatineforamen from where nasopharyngeal angi<strong>of</strong>ibromasoriginate. This <strong>the</strong>ory explains <strong>the</strong> abnormal vascularstructures <strong>and</strong> <strong>the</strong> complex anatomical extensions<strong>of</strong> nasopharyngeal angi<strong>of</strong>ibromas. The growth stimulationduring puberty <strong>and</strong> <strong>the</strong> restriction to males remainunexplained by this <strong>the</strong>ory, however. Treatment <strong>of</strong> nasopharyngealangi<strong>of</strong>ibromas is surgical after embolisation,although radiation <strong>the</strong>rapy <strong>and</strong> hormonal <strong>the</strong>rapyhave been used extensively in <strong>the</strong> past [15, 37]. Nasopharyngealangi<strong>of</strong>ibromas have no malignant potential, exceptfor radiation-related malignant transformation.6.2.4.2 Respiratory Epi<strong>the</strong>lialAdenomatoid HamartomaRespiratory epi<strong>the</strong>lial adenomatoid hamartoma (or polypoidhamartomas) typically arises within <strong>the</strong> nasal cavity<strong>and</strong> paranasal sinuses (see also Chap. 2), but has alsobeen reported in <strong>the</strong> nasopharynx [199]. Patients areadults with an age range <strong>of</strong> 24–81 years. The polypoidexophytic hamartomas are rubbery, tan to brown <strong>and</strong>can reach up to 6 cm in size. They are lined with ciliatedrespiratory epi<strong>the</strong>lium with mucin-secreting gobletcells. The widely spaced gl<strong>and</strong>ular proliferations arisefrom invagination <strong>of</strong> <strong>the</strong> surface epi<strong>the</strong>lium. A thickeosinophilic basement membrane surrounds <strong>the</strong> gl<strong>and</strong>s<strong>and</strong> surface epi<strong>the</strong>lium. The ample stroma may be oedematous<strong>and</strong> well-vascularised or fibrous with varyingamounts <strong>of</strong> lymphocytes <strong>and</strong> inflammatory cells(Fig. 6.4). Gl<strong>and</strong>ular acinar proliferations may be scant<strong>and</strong> large cysts may predominate when <strong>the</strong> fibrous stromapredominates [206]. Some nasopharyngeal hamartomasinclude a chondro-osseous component <strong>and</strong> cystslined with squamous epi<strong>the</strong>lium. Due to overgrowth <strong>of</strong>a single mesenchymal element, respiratory epi<strong>the</strong>lialadenomatoid hamartomas may resemble fibromas, lipomasor chondromas [199]. Treatment <strong>of</strong> choice is surgery.The differential diagnoses <strong>of</strong> respiratory epi<strong>the</strong>lialadenomatoid hamartoma include inverted papilloma<strong>and</strong> adenocarcinoma. The presence <strong>of</strong> excess gl<strong>and</strong>s <strong>and</strong><strong>the</strong> respiratory epi<strong>the</strong>lium distinguish <strong>the</strong> hamartomareadily from an inverted papilloma. The lack <strong>of</strong> malignantcytological <strong>and</strong> histological features <strong>and</strong> invasiondistinguishes <strong>the</strong> respiratory epi<strong>the</strong>lial adenomatoidhamartoma from an adenocarcinoma.6.2.4.3 NasopharyngealInverted PapillomaICD-O:8121/1Inverted papillomas (or Schneiderian papillomas) arisingoutside <strong>the</strong> sinonasal tract are extremely rare. Onepublication reported 15 pharyngeal inverted papillomas,11 <strong>of</strong> which arose in <strong>the</strong> nasopharynx [183]. The pink orgrey firm polyps had a convoluted surface, which cor-


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 179papillomas have an increased potential for recurrence,which is higher in <strong>the</strong> nasopharynx than in <strong>the</strong> sinonasallocation. This probably reflects <strong>the</strong> high rate <strong>of</strong> incompleteinitial removal due to <strong>the</strong> unexpected nature <strong>of</strong> <strong>the</strong>lesion <strong>and</strong> <strong>the</strong> difficult nasopharyngeal anatomy. As ina sinonasal location, inverted papillomas are at risk <strong>of</strong>malignant transformation. For a detailed description <strong>of</strong>inverted papillomas, see Chap. 2.6.2.4.4 Solitary Fibrous TumouraICD-O:8815/0Solitary fibrous tumours are exceptionally rare in <strong>the</strong>upper respiratory tract. In <strong>the</strong> nasopharynx, only 4 caseshave been described to date [96, 122, 201].6.2.4.5 ParagangliomabICD-O:8680/1Paraganglioma (or chemodectoma) arise in <strong>the</strong> head <strong>and</strong>neck region most commonly in <strong>the</strong> carotid body, glomustympanicus <strong>and</strong> glomus jugulare. They are extremelyrare in <strong>the</strong> nasopharynx with about 20 cases described(for review see [101]), but need to be considered whenpr<strong>of</strong>use haemorrhage <strong>and</strong> a pulsatile mass are encounteredin <strong>the</strong> nasopharynx. These slowly enlarging, painless,firm, encapsulated tumours are characterised bynests (so-called Zellballen) <strong>of</strong> small to medium-sizedpale monomorphous cells with prominent, roundedhyperchromatic nuclei <strong>and</strong> a prominent network <strong>of</strong> endo<strong>the</strong>lialcell lined vascular channels (see also Chaps. 8<strong>and</strong> 9).6.2.4.6 MeningiomacFig. 6.4. Respiratory epi<strong>the</strong>lial adenomatoid hamartoma. a Thisfragmented surgical specimen <strong>of</strong> polypoid hamartoma displaysnumerous fluid <strong>and</strong> mucin filled cysts. b The surface is lined bytall columnar respiratory epi<strong>the</strong>lium <strong>and</strong> <strong>the</strong>re are widely spacedacinar proliferations embedded in a fibrous stroma. c The acinarproliferations are in continuity with invaginations <strong>of</strong> <strong>the</strong> surfacerespiratory epi<strong>the</strong>lium <strong>and</strong> recapitulate ei<strong>the</strong>r seromucous gl<strong>and</strong>s.The basement membrane is typically thickenedresponded histologically to a multilayered, transitionaltypeepi<strong>the</strong>lium with foci <strong>of</strong> respiratory <strong>and</strong> squamousepi<strong>the</strong>lium growing endophytically into <strong>the</strong> underlyingloose <strong>and</strong> myxoid stroma. Nasopharyngeal invertedICD-O:9530/0Among <strong>the</strong> rarest tumours <strong>of</strong> <strong>the</strong> nasopharynx are extracranialmeningiomas [162]. In a series <strong>of</strong> 30 extracranialmeningiomas, 3 were located in <strong>the</strong> nasopharynx[188]. There is only a single case report <strong>of</strong> a primary tonsillarectopic meningioma [107]. Extracranial meningiomasshow <strong>the</strong> same histological subtypes, differentiation<strong>and</strong> prognosis as intracranial meningiomas, but <strong>the</strong> syncytial(meningo<strong>the</strong>lial) subtype predominates. For meningiomasat o<strong>the</strong>r sites in <strong>the</strong> head <strong>and</strong> neck area, seeChaps. 2, 8 <strong>and</strong> 10.6.2.4.7 Gl<strong>and</strong>ular Retention CystsGl<strong>and</strong>ular retention cysts are ra<strong>the</strong>r common in <strong>the</strong> oro<strong>and</strong>nasopharynx <strong>and</strong> some may become large enoughto mimic nasopharyngeal tumours. They arise from di-


180 S. Regauer6lated <strong>and</strong> occluded excretory ducts <strong>of</strong> <strong>the</strong> seromucinoussalivary gl<strong>and</strong>s <strong>of</strong> <strong>the</strong> nasopharynx. The cysts are filledwith mucin <strong>and</strong> lined with a single layer <strong>of</strong> cuboidalepi<strong>the</strong>lium. Varying amounts <strong>of</strong> squamous metaplasia<strong>and</strong> oncocytic metaplasia can be observed, <strong>and</strong> whenextensive, <strong>the</strong> designation oncocytic cyst or nasopharyngealoncocytoma may be most appropriate [3, 12, 136].Retention cysts may rupture. The ensuing inflammationinduced periductal fibrosis <strong>and</strong> an inflammatoryinfiltrate. The epi<strong>the</strong>lium is destroyed <strong>and</strong> replaced bymacrophages. These mucin-filled cysts are <strong>the</strong>n calledmucoceles.6.2.5 Nasopharyngeal CarcinomaClassification <strong>of</strong> nasopharyngeal carcinomas has undergoneseveral changes in <strong>the</strong> past editions <strong>of</strong> <strong>the</strong> WHOclassification. The 1991 edition separates nasopharyngealcarcinoma into two subtypes, <strong>the</strong> keratinising <strong>and</strong><strong>the</strong> non-keratinising carcinoma [176]. The non-keratinisingcarcinomas have been fur<strong>the</strong>r subdivided intoundifferentiated <strong>and</strong> differentiated forms. This subdivision,however, is nowadays considered optional since<strong>the</strong>ir distinction is <strong>of</strong> no clinical or prognostic significance.Whe<strong>the</strong>r basaloid-squamous carcinoma meritsseparate recognition or merely represents a morphologicvariant <strong>of</strong> non-keratinising carcinoma has not yet beenclarified [7, 138, 159, 194].6.2.5.1 Non-KeratinisingNasopharyngeal CarcinomaIn <strong>the</strong> past, non-keratinising nasopharyngeal carcinomashave been divided into <strong>the</strong> more common undifferentiatednasopharyngeal carcinoma (ICD-O:8082/3),(synonymous lymphoepi<strong>the</strong>lial carcinoma, lymphoepi<strong>the</strong>lioma<strong>of</strong> Regaud <strong>and</strong> Schmincke, undifferentiated carcinomawith lymphoid stroma) <strong>and</strong> <strong>the</strong> less common differentiatednasopharyngeal carcinoma (ICD-O:8073/3)(or transitional-type carcinoma, intermediate cell carcinoma).The distinction between undifferentiated <strong>and</strong>differentiated carcinoma is considered unimportant for<strong>the</strong>rapy <strong>and</strong> prognosis, as <strong>the</strong>se two subtypes representa spectrum <strong>of</strong> <strong>the</strong> same tumour. Nasopharyngeal carcinomais by far <strong>the</strong> most common carcinoma in China<strong>and</strong> Taiwan, accounting for 18% <strong>of</strong> all carcinomas <strong>the</strong>re,with an incidence <strong>of</strong> 10–20/100,000 [83]. Nasopharyngealcarcinomas also occur in endemic forms in Asia,Greenl<strong>and</strong>, Alaska <strong>and</strong> Africa. In <strong>the</strong>se high-incidenceareas, 99% <strong>of</strong> nasopharyngeal carcinomas are <strong>of</strong> <strong>the</strong>non-keratinising subtype. In <strong>the</strong> western world – a lowincidencearea with 0.4–1/100,000 – <strong>the</strong> non-keratinisingnasopharyngeal carcinoma accounts for 75% comparedwith 25% for keratinising carcinoma [8, 43, 141]. Nasopharyngealcarcinomas show a strong male predilection.While most patients with non-keratinising nasopharyngealcarcinoma are older than 50 years in endemic areas,<strong>the</strong>re is a bimodal age distribution with a peak presentationin <strong>the</strong> 2nd <strong>and</strong> 6th decades in intermediate- <strong>and</strong>low-incidence areas [8, 43, 63, 77, 175]. The causative <strong>and</strong>aetiological role <strong>of</strong> EBV is well established in invasive<strong>and</strong> in situ nasopharyngeal carcinomas, irrespective <strong>of</strong><strong>the</strong> ethnic origin <strong>of</strong> <strong>the</strong> patient <strong>and</strong> <strong>the</strong> histological subtype[31, 158]. However, environmental factors seem toplay a role, since <strong>the</strong> incidence <strong>of</strong> non-keratinising nasopharyngealcarcinoma decreases among second <strong>and</strong>third generation Chinese living in non-endemic areas[83]. Nasopharyngeal carcinomas arise in <strong>the</strong> lateralwalls <strong>of</strong> <strong>the</strong> nasopharynx in <strong>the</strong> area <strong>of</strong> <strong>the</strong> Rosenmüllerfossa <strong>and</strong> presenting symptoms may be nasal obstruction,epistaxis, post-nasal drip, tinnitus <strong>and</strong> cranialnerve palsy. Hearing loss <strong>and</strong> unilateral otitis mediaare related to auditory tube involvement. In more than50% <strong>of</strong> patients, however, metastases to cervical lymphnodes are <strong>the</strong> presenting sign. Since most nasopharyngealcarcinomas are difficult to visualise on endoscopicexamination, “blind” biopsies <strong>of</strong> <strong>the</strong> nasopharynx, base<strong>of</strong> tongue <strong>and</strong> palatine tonsils are necessary to establisha histological diagnosis <strong>of</strong> undifferentiated nasopharyngealcarcinoma. In patients presenting with lymphnode metastases <strong>and</strong> clinically occult primary carcinoma,demonstration <strong>of</strong> EBV in <strong>the</strong> metastatic carcinomamay be a helpful diagnostic tool to correctly identify <strong>the</strong>primary nasopharyngeal carcinoma [26, 40, 119, 191].Nasopharyngeal carcinomas are highly responsive toradiation <strong>the</strong>rapy. The overall 5-year survival has beenreported to be between 25 <strong>and</strong> 50% in <strong>the</strong> past, but <strong>the</strong>results <strong>of</strong> treatment have improved due to refinementsin staging <strong>and</strong> techniques <strong>of</strong> <strong>the</strong>rapy [4, 115].Histologically, non-keratinising nasopharyngeal carcinomaslack gl<strong>and</strong>ular differentiation. The undifferentiatedsubtype consists <strong>of</strong> bl<strong>and</strong> uniform undifferentiatedcells in large cohesive nests <strong>and</strong> cords as well as smallernests <strong>and</strong> groups <strong>of</strong> epi<strong>the</strong>lial cells with numerous mitoses.They may be sharply outlined <strong>and</strong> separated from<strong>the</strong> surrounding infiltrate or permeated by a dense inflammatoryinfiltrate consisting <strong>of</strong> numerous lymphocytes,plasma cells <strong>and</strong> eosinophilic granulocytes. Thispattern <strong>of</strong>ten has a distinctly non-carcinomatous <strong>and</strong>lymphoma-like appearance. The infiltrating lymphocytesare a mixture <strong>of</strong> plasma cells, B-cells <strong>and</strong> T-cells,with B-cells usually predominating. Portions <strong>of</strong> <strong>the</strong> T-cells are cytotoxic cells. A variant <strong>of</strong> undifferentiatedcarcinoma shows extensive acantholysis resulting in apseudogl<strong>and</strong>ular <strong>and</strong> pseudovascular pattern. The differentiatedsubtype shows cellular stratification <strong>of</strong>ten ina plexiform growth, reminiscent <strong>of</strong> transitional cell carcinoma<strong>of</strong> <strong>the</strong> urinary bladder (Fig. 6.5). The stroma <strong>of</strong>non-keratinising nasopharyngeal carcinoma can be fibrous,but is rarely desmoplastic. Coagulative necrosis


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 181abcdefFig. 6.5. Non-keratinizing nasopharyngeal carcinoma. a–b Nasopharyngealcarcinoma grows in solid sheets <strong>of</strong> large epi<strong>the</strong>lialcells which are clearly demarcated by a mixed inflammatory infiltrateconsisting <strong>of</strong> lymphocytes, plasma cells <strong>and</strong> histiocytes.c globular amyloid deposition is not uncommon. d–e Non-keratinizingnasopharyngeal carcinoma can be heavily infiltrated by<strong>the</strong> lymphocytes. f Tumour cells are large, undifferentiated, withabundant pale cytoplasms, inconspicuous cell membranes, <strong>and</strong>large nuclei with prominent nucleolimay be present. Small amyloid globules can be foundwithin <strong>the</strong> tumour cells or scattered among <strong>the</strong> carcinomatousstroma. Epi<strong>the</strong>lioid granulomas, occasionallywith caseous necrosis, have been described in nasopharyngealcarcinoma in up to 18% <strong>of</strong> cases. Some featuresare associated with a better prognosis <strong>of</strong> nasopharyngealcarcinoma: high density <strong>of</strong> dendritic cells, highnumber <strong>of</strong> infiltrating lymphocytes, <strong>and</strong> low numbers<strong>of</strong> granzyme-B positive cytotoxic cells [58, 80, 151, 208].The most reliable <strong>and</strong> sensitive method <strong>of</strong> detection <strong>of</strong>


182 S. Regauer6EBV in paraffin sections in routine diagnostic pathologyis EBER in situ hybridisation [88, 93, 157, 196]. Themain differential diagnosis <strong>of</strong> nasopharyngeal carcinomais a non-Hodgkin’s lymphoma, particularly <strong>the</strong> largecell lymphoma. The diagnosis <strong>of</strong> a nasopharyngeal carcinomacan be confirmed by positive staining with antibodiesto cytokeratins, especially high molecular weightkeratin.6.2.5.2 KeratinisingNasopharyngeal CarcinomaICD-O:8070/3Keratinising nasopharyngeal carcinoma (squamous cellcarcinoma, SCC) occurs typically after 40 years <strong>of</strong> age<strong>and</strong> shows obvious squamous differentiation with varyingamounts <strong>of</strong> keratinisation. The stroma is desmoplastic<strong>and</strong> infiltrated by variable numbers <strong>of</strong> lymphocytes,plasma cells, neutrophils <strong>and</strong> eosinophils. Poorly differentiatedSCC may only contain rare horn pearls orfocal areas <strong>of</strong> easily recognisable cornification. Immunohistochemicalanalysis with antibody to involucrinis helpful in identifying areas <strong>of</strong> abortive keratinisation[98]. EBV is almost always positive in nasopharyngealSCC in endemic areas for nasopharyngeal carcinoma,but only a small number <strong>of</strong> cases in low incidence areasare positive for EBV. SCC shows a greater propensityfor localised advanced tumour growth, but a lower rate<strong>of</strong> lymph node metastases [141, 164]. Radical surgery isnot performed since radio<strong>the</strong>rapy is extremely effective.Survival <strong>and</strong> prognosis <strong>of</strong> SCC depends on tumour stage<strong>and</strong> has been reported to be better than that for non-keratinisingnasopharyngeal carcinoma.6.2.6 NasopharyngealAdenocarcinomaAdenocarcinomas <strong>of</strong> <strong>the</strong> nasopharynx are extremelyuncommon tumours <strong>and</strong> can be separated into carcinomasarising from <strong>the</strong> surface epi<strong>the</strong>lium or from <strong>the</strong>mucoserous salivary gl<strong>and</strong>s. The salivary gl<strong>and</strong>-typecarcinomas are more common than those arising from<strong>the</strong> surface epi<strong>the</strong>lium [114].6.2.6.1 Salivary Gl<strong>and</strong>-TypeAdenocarcinoma<strong>of</strong> <strong>the</strong> NasopharynxAdenoid cystic carcinoma (ICD-O:8200/3), <strong>the</strong> mostcommon carcinoma <strong>of</strong> <strong>the</strong> salivary type in <strong>the</strong> oral cavity<strong>and</strong> hard palate, is rare in <strong>the</strong> nasopharynx [21, 114,181]. Adenoid cystic carcinomas <strong>of</strong> <strong>the</strong> nasopharynx aremore common in Japan than in <strong>the</strong> western world. It isan insidiously growing tumour with a predispositionfor perineural spread, local recurrence <strong>and</strong> distant metastasis.Regional lymph node metastases are rare. Histologically,adenoid cystic carcinomas are classified intubular, cribriform <strong>and</strong> solid subtypes. The tubular <strong>and</strong>cribriform subtypes are considered low-grade tumours;<strong>the</strong> solid sub-type is a high-grade tumour with a rapid,fatal course <strong>and</strong> a higher incidence <strong>of</strong> distant metastasiswith a poor prognosis [114]. When compared withconventional nasopharyngeal carcinoma, adenoid cysticcarcinoma has a higher incidence <strong>of</strong> cranial nerve involvement,but a lower incidence <strong>of</strong> cervical lymph nodemetastases.Polymorphous low-grade adenocarcinoma <strong>of</strong> minorsalivary gl<strong>and</strong>s (ICD-O:8525/3) (or terminal ductcarcinoma, lobular carcinoma, low-grade papillary adenocarcinoma)is a low-grade neoplasm typically occurringin <strong>the</strong> oral cavity. It has been documented in<strong>the</strong> nasopharynx in rare cases [144, 198]. The polymorphouslow-grade carcinoma has a wide diversity<strong>of</strong> histological patterns including solid areas, papillarygrowth, ductal differentiation, cystic spaces <strong>and</strong> an infiltrativegrowth pattern with perineural invasion. Themain bulk <strong>of</strong> <strong>the</strong> carcinoma is found in <strong>the</strong> submucosa<strong>and</strong> <strong>the</strong> surface epi<strong>the</strong>lium is <strong>of</strong>ten intact. In <strong>the</strong> nasopharynx,surgery or radio<strong>the</strong>rapy is <strong>the</strong> treatment <strong>of</strong>choice. Polymorphous low-grade carcinomas <strong>of</strong> salivarygl<strong>and</strong> origin have a potentially aggressive biologicalcourse with metastases to cervical lymph nodes.For a detailed description <strong>of</strong> salivary gl<strong>and</strong> tumourssee Chap. 5.6.2.6.2 Papillary Adenocarcinoma<strong>of</strong> <strong>the</strong> NasopharynxICD-O:8260/3Papillary adenocarcinomas <strong>of</strong> <strong>the</strong> nasopharynx are extremelyuncommon, slow-growing, low-grade carcinomaswithout known risk factors [76, 114]. In a series <strong>of</strong>9 patients, aged 11 to 64 years, <strong>the</strong> papillary adenocarcinomas<strong>of</strong> <strong>the</strong> nasopharynx were confined to <strong>the</strong> ro<strong>of</strong><strong>and</strong> postero-lateral walls <strong>of</strong> <strong>the</strong> nasopharynx [198]. Papillarynasopharyngeal adenocarcinomas are unencapsulated,exophytic, cauliflower-like tumours with an occasionalgritty consistency due to numerous psammomabodies. Complex papillations arise from <strong>the</strong> surface epi<strong>the</strong>lium.The papillae <strong>and</strong> crowded back-to-back gl<strong>and</strong>sare lined with uniform, bl<strong>and</strong>, tall, columnar cells withintermixed mucin-containing, PAS-positive goblet cells.Some areas show overlapping vesicular nuclei with granularcytoplasm resembling thyroid carcinomas. Stromalcalcifications <strong>and</strong> psammoma bodies can be found. Vascular,lymphatic or neural invasion is uncommon. Lowgradepapillary adenocarcinomas <strong>of</strong> <strong>the</strong> nasopharynxshould be distinguished from <strong>the</strong> polymorphous low-


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 183grade carcinoma <strong>of</strong> salivary gl<strong>and</strong> origin because <strong>the</strong>ydo not metastasise.6.2.7 Malignant Non-Epi<strong>the</strong>lialTumours <strong>of</strong> <strong>the</strong> Nasopharynx6.2.7.1 ChordomaICD-O:9370/3Chordomas are tumours arising from remnants <strong>of</strong> <strong>the</strong>notochord in <strong>the</strong> axial skeleton near its cranial <strong>and</strong> caudalends. The cranially located chordomas compriseabout one-third <strong>of</strong> all chordomas, arise earlier than <strong>the</strong>sacral chordomas, around <strong>the</strong> 3rd to <strong>the</strong> 5th decade <strong>and</strong>in children. Most chordomas in nasopharyngeal locationare extensions <strong>of</strong> cranial chordomas, but <strong>the</strong>y rarelyarise de novo in <strong>the</strong> nasopharynx <strong>and</strong> paranasal sinuses[19, 34, 54]. For a more detailed description <strong>of</strong> chordoma,see Chap. 4.6.2.7.2 SarcomaThe most common sarcoma in <strong>the</strong> head <strong>and</strong> neck areais <strong>the</strong> rhabdomyosarcoma, specifically <strong>the</strong> embryonalrhabdomyosarcoma (ICD-O:8910/3) in children lessthan 5 years <strong>of</strong> age [49]. Primary nasopharyngeal rhabdomyosarcomasare rare [20, 39, 50] as are primary nasopharyngealchondrosarcomas (ICD-O:9220/3) [54,56]. Case reports <strong>of</strong> primary nasopharyngeal sarcomasinclude an osteosarcoma in an 11-year-old girl aftermulti-agent chemo<strong>the</strong>rapy <strong>and</strong> radiation treatment <strong>of</strong> aretinoblastoma, a liposarcoma <strong>and</strong> a granulocytic sarcomain a 37-year-old Chinese man [5, 42, 139]. The folliculardendritic cell sarcoma, a tumour <strong>of</strong> antigen-presentingcells <strong>of</strong> B-follicles <strong>of</strong> lymphoid tissues, is commonlyfound extranodally in <strong>the</strong> head <strong>and</strong> neck area, butrarely in <strong>the</strong> nasopharynx [10, 22, 23]. It is characterisedby positivity for CD21, CD35 <strong>and</strong> CD23, as well as indolentclinical behaviour <strong>and</strong> a low risk <strong>of</strong> recurrence <strong>and</strong>metastasis [87].6.3 Waldeyer’s Ring6.3.1 Anatomy <strong>and</strong> Histology<strong>of</strong> Waldeyer’s RingThe term Waldeyer’s ring refers to <strong>the</strong> ring <strong>of</strong> lymphoidtissues occurring in <strong>the</strong> nasopharynx <strong>and</strong> oropharynx.The oropharynx is separated from <strong>the</strong> nasopharynxby <strong>the</strong> oropharyngeal isthmus, which is formed by <strong>the</strong>merging muscular pillars <strong>of</strong> <strong>the</strong> palatoglossal <strong>and</strong> <strong>the</strong>palatopharyngeal muscles. In <strong>the</strong> lateral walls, at <strong>the</strong>widest points <strong>of</strong> <strong>the</strong> pharynx, lies <strong>the</strong> triangular tonsillarfossa, which contains <strong>the</strong> palatine tonsil. The tonsils<strong>of</strong> Waldeyer’s ring belong to <strong>the</strong> gut-associated lymphoepi<strong>the</strong>lialorgans, which show a close morphological <strong>and</strong>functional correlation between lymphatic tissue <strong>of</strong> mesenchymalorigin <strong>and</strong> <strong>the</strong> endodermal epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong>second pharyngeal pouch. The Waldeyer’s ring tonsilsare composed <strong>of</strong> <strong>the</strong> paired palatine tonsils in <strong>the</strong> tonsillarfossa, <strong>the</strong> unpaired (naso)pharyngeal tonsil in <strong>the</strong>ro<strong>of</strong> <strong>of</strong> <strong>the</strong> nasopharynx, <strong>the</strong> bilateral tubal tonsils in <strong>the</strong>lateral walls <strong>of</strong> <strong>the</strong> nasopharynx at <strong>the</strong> entrance to <strong>the</strong>auditory tube <strong>and</strong> <strong>the</strong> lingual tonsil in <strong>the</strong> retrolingualregion. Lymphoid aggregations close to <strong>the</strong> epiglottis arealso counted as part <strong>of</strong> Waldeyer’s ring.The palatine tonsils are <strong>the</strong> largest <strong>of</strong> <strong>the</strong> tonsils <strong>and</strong>lie in <strong>the</strong> tonsillar fossa along <strong>the</strong> anterolateral border<strong>of</strong> <strong>the</strong> oropharynx between <strong>the</strong> palatoglossal <strong>and</strong> palatopharyngealmuscle arches. The lateral surface <strong>of</strong> <strong>the</strong>palatine tonsil has a fibrous capsule, but skeletal musclefibres <strong>and</strong> isl<strong>and</strong>s <strong>of</strong> mostly elastic cartilage are normalfindings <strong>the</strong>re. The medial surface has 10–30 epi<strong>the</strong>lium-lined,extensively branching <strong>and</strong> anastomosing tonsillarcrypts that extend deeply into <strong>the</strong> lymphoid tissue.The surface <strong>of</strong> <strong>the</strong> palatine tonsils is lined by stratifiedsquamous epi<strong>the</strong>lium, <strong>the</strong> crypts are covered by nonkeratinisingstratified (“transitional-type”) epi<strong>the</strong>liumwith a discontinuous basement membrane <strong>and</strong> numerousintraepi<strong>the</strong>lial lymphoid cells. This so-called lymphoepi<strong>the</strong>liumrepresents <strong>the</strong> specialised epi<strong>the</strong>lium <strong>of</strong><strong>the</strong> tonsils <strong>and</strong> contains M-cells (resembling <strong>the</strong> intestinalmembranous [M] cells <strong>of</strong> <strong>the</strong> Peyer’s patches), T- <strong>and</strong>B-cells, <strong>and</strong> patchily distributed macrophages <strong>and</strong> dendriticcells [140, 152]. The surface epi<strong>the</strong>lium is evenlyinfiltrated by T-cells <strong>and</strong> B-cells; up to 30% <strong>of</strong> <strong>the</strong> intraepi<strong>the</strong>lialT-cells are T-cells, which are involved inantigen recognition independent <strong>of</strong> MHC restriction<strong>and</strong> prior antigen processing [66, 149]. A clustering <strong>of</strong>CD4+ T-cells with B-cells is typical within <strong>the</strong> lymphoepi<strong>the</strong>lium<strong>of</strong> <strong>the</strong> crypts <strong>and</strong> in <strong>the</strong> submucosa [66]. Thesubmucosal lymphoid tissue contains numerous primary<strong>and</strong> secondary lymph follicles with germinal centres,a mantle zone <strong>and</strong> a network <strong>of</strong> follicular dendritic cells.T-cells, interdigitating dendritic cells, plasma cells, macrophages<strong>and</strong> high-endo<strong>the</strong>lial venules are found in <strong>the</strong>extrafollicular regions [140]. The palatine tonsils haveno afferent lymph vessels. The rich efferent lymphaticdrainage is via <strong>the</strong> retropharyngeal lymph nodes to <strong>the</strong>upper deep cervical lymph nodes.The small unencapsulated (naso)pharyngeal tonsilwith about 12–15 shallow crypts is lined with a columnarciliated respiratory surface epi<strong>the</strong>lium with numerousgoblet cells, which may also be seen in <strong>the</strong> lymphoepi<strong>the</strong>liallining <strong>of</strong> <strong>the</strong> short <strong>and</strong> plump crypts [200]. Thelymphoid tissue contains numerous lymph follicles withgerminal centres. Minor salivary gl<strong>and</strong>s in <strong>the</strong> periph-


184 S. Regauer6ery <strong>and</strong> within <strong>the</strong> submucosa <strong>of</strong> <strong>the</strong> pharyngeal tonsilare a normal finding. The tubal tonsils are poorly definedcondensations <strong>of</strong> lymphoid tissue located around<strong>the</strong> auditory tube in <strong>the</strong> nasopharynx <strong>and</strong> around <strong>the</strong>fossa <strong>of</strong> Rosenmüller. The term lingual tonsil refers to <strong>the</strong>abundant non-encapsulated lymphoid tissue in <strong>the</strong> adultposterior tongue. The crypts do not appear until birth,are shallow <strong>and</strong> much less branched than in <strong>the</strong> palatinetonsil. The surface is covered by non-keratinising squamousepi<strong>the</strong>lium. Adipose tissue <strong>and</strong> skeletal muscle fibresat <strong>the</strong> base <strong>of</strong> <strong>the</strong> lingual tonsil, <strong>and</strong> mucous salivarygl<strong>and</strong>s <strong>and</strong> lymph follicles within <strong>the</strong> lingual tonsilare integral parts <strong>of</strong> <strong>the</strong> tongue. Efferent lymphaticvessels <strong>of</strong> <strong>the</strong> posterior tongue drain into <strong>the</strong> deep cervicallymph nodes, after <strong>the</strong>y pass through <strong>the</strong> pharyngealwall in front <strong>of</strong> or behind <strong>the</strong> external carotid artery[41].6.3.2 Congenital Anomalies<strong>of</strong> Waldeyer’s RingCongenital anomalies include <strong>the</strong> extremely rare absence<strong>of</strong> palatine tonsils <strong>and</strong> accessory tonsils within <strong>the</strong>oral cavity [68, 155]. Slightly more common is <strong>the</strong> “hairypolyp”, a choristoma, which arises from remnants <strong>of</strong> <strong>the</strong>ectodermal <strong>and</strong> mesodermal germ layers in <strong>the</strong> palatinetonsil <strong>and</strong> nasopharynx (see also Sect. 6.2.3.2). The unilateralpedunculated hairy polyps <strong>of</strong> up to 5 cm arisemostly on <strong>the</strong> superior pole <strong>of</strong> <strong>the</strong> palatine tonsil <strong>and</strong>present with acute respiratory distress [129]. A rare bilateralpresentation has been described [52]. These polypsshow a mesenchymal fibrovascular core, <strong>of</strong>ten withadipose tissue <strong>and</strong> skeletal muscle fibres <strong>and</strong> are coveredin regular skin with hair appendages. In tonsillar hairypolyps, submucosal plates <strong>of</strong> elastic cartilage <strong>of</strong> uniformthickness resembling <strong>the</strong> external ear auricular tags <strong>and</strong>auricular cartilage have been reported [78]. Hairy polyps<strong>of</strong> <strong>the</strong> tonsils have been postulated to arise from <strong>the</strong>second pharyngeal pouch [17], but due to <strong>the</strong> presence<strong>of</strong> <strong>the</strong> cartilage plates <strong>the</strong>y have also been interpreted asaccessory auricles with a postulated origin from <strong>the</strong> firstpharyngeal arch [78].6.3.3 TonsillitisThe lymphoid tissues <strong>of</strong> Waldeyer’s ring play a key rolein initiating immune responses against inhaled <strong>and</strong>ingested pathogens. The tonsils are responsible for <strong>the</strong>recognition <strong>and</strong> processing <strong>of</strong> antigens presented to <strong>the</strong>pharynx. The size <strong>of</strong> <strong>the</strong> tonsils is directly proportionalto <strong>the</strong> amount <strong>of</strong> lymphoid tissue, which increases duringantigen challenge. The reactive lymphoid hyperplasia<strong>of</strong> <strong>the</strong> palatine tonsils is <strong>of</strong>ten simply referred to as“tonsillitis” <strong>and</strong> in <strong>the</strong> case <strong>of</strong> <strong>the</strong> pharyngeal tonsil as“(hyperplastic) adenoids”. Tonsillar hypertrophy is associatedwith normal childhood development, mostlydue to viral challenge or can be secondary to specificbacterial or viral infections. Childhood hypertrophy <strong>of</strong><strong>the</strong> pharyngeal tonsil begins at approximately 2 years <strong>of</strong>age or during infancy <strong>and</strong> usually regresses by 8 years <strong>of</strong>age. Palatine tonsils hypertrophy at <strong>the</strong> end <strong>of</strong> <strong>the</strong> firstdecade, somewhat later than <strong>the</strong> pharyngeal tonsil. Theyregress by puberty <strong>and</strong> are atrophied in adults. The lingualtonsil enlarges at <strong>the</strong> time <strong>of</strong> puberty <strong>and</strong> regressesvery little during adult life [41]. Tonsillar hypertrophyis usually symmetrical <strong>and</strong> diffuse, but can be papillary<strong>and</strong> unilateral.The normal flora <strong>of</strong> <strong>the</strong> naso- <strong>and</strong> oropharynx includesanaerobic bacteria such as gram-positive Actinomyces<strong>and</strong> Proprionibacterium, <strong>and</strong> gram-negativebacteria such as Bacteroides, Fusobacterium <strong>and</strong> Vibrio[202]. Actinomyces israelii is a common nosocomialsaprophyte in <strong>the</strong> oro-/nasopharyngeal cavity. The trueincidence <strong>of</strong> tonsillar manifestations <strong>of</strong> actinomyces isunknown, but has been reported to be as high as 40%[57, 120]. Occasionally, actinomyces form small sulphurgranules that can be seen as small yellow dots on <strong>the</strong>tonsillar surface. Larger aggregates <strong>of</strong> actinomyces mayproduce a tumour-like mass [172]. The tangled masses<strong>of</strong> gram-positive branching mycelial-like bacteria liewithin <strong>the</strong> crypts or are attached to <strong>the</strong> surface epi<strong>the</strong>lium<strong>of</strong> normal tonsils. Tonsillectomies for hypertrophiedtonsils or adenoids are one <strong>of</strong> <strong>the</strong> most common surgicalprocedures, but <strong>the</strong> term “tonsillitis” is still poorlydefined. Surgical resection specimens may demonstrateonly hyperplastic lymphoid tissue <strong>and</strong> lymph follicleswith enlarged germinal centres or no pathology atall [91].6.3.3.1 Bacterial TonsillitisBacterial suppurative tonsillitis is among <strong>the</strong> most frequentpaediatric infections. Group A beta-haemolyticstreptococci are <strong>the</strong> most frequent cause. O<strong>the</strong>r commonisolates in bacterial tonsillitis are Hemophilus influenza,Streptococcus pyogenes, Streptococcus milleri <strong>and</strong> Staphylococcusaureus [97, 202, 205]. Children with acute streptococcaltonsillitis are significantly older than childrenwith viral tonsillitis. The treatment <strong>of</strong> choice is penicillinadministration for 10 days. Prevention <strong>of</strong> acute rheumaticfever is <strong>the</strong> principal goal <strong>of</strong> treatment. Surgicalspecimens <strong>of</strong> acute tonsillitis are rarely encountered. Thesurface epi<strong>the</strong>lium may be ulcerated, <strong>and</strong> <strong>the</strong> surface <strong>and</strong>crypt epi<strong>the</strong>lium is infiltrated by neutrophilic granulocytesproducing a cryptitis with crypt abscesses. Acutebacterial infections may advance to intraparenchymal<strong>and</strong> peritonsillar abscesses (quinsy) with a lateral extensioninto <strong>the</strong> parapharyngeal space, base <strong>of</strong> skull <strong>and</strong> <strong>the</strong>sheath <strong>of</strong> <strong>the</strong> carotid artery [33, 64]. Rare o<strong>the</strong>r bacteria


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 185causing acute necrotising tonsillitis include Clostridiumperfringens <strong>and</strong> Bartonella henselae with an unusual presentation<strong>of</strong> cat scratch disease [61, 121].6.3.3.2 Viral TonsillitisThe most common causes <strong>of</strong> upper respiratory tractinfections <strong>and</strong> pharyngo-tonsillitis in <strong>the</strong> general population,including infants <strong>and</strong> young children, are virusessuch as influenza virus, Coxsackie’s virus (groupA), adenovirus, <strong>and</strong> <strong>the</strong> ubiquitous herpes virus Epstein-Barrvirus [205]. EBV infects epi<strong>the</strong>lial cells <strong>and</strong>B-lymphocytes <strong>of</strong> Waldeyer’s ring, which represent<strong>the</strong> reservoir for life-long viral persistence [104, 186].Primary infections with EBV occur early in infancy<strong>and</strong> childhood in developing countries <strong>and</strong> are generallyasymptomatic. In contrast, in developed countries,primary infection occurs in adolescents <strong>and</strong> youngadults. EBV infections may cause <strong>the</strong> mostly self-limitingacute disease infectious mononucleosis, affectingadolescents <strong>and</strong> young adults in <strong>the</strong> western world. InJapan, however, an endemic area for EBV, acute cases <strong>of</strong>infectious mononucleosis are commonly diagnosed inchildren less than 4 years <strong>of</strong> age [100]. The symptomsinclude enlarged swollen palatine tonsils, occasionallywith peritonsillar abscesses, sore throat, fever, malaise,cervical lymphadenopathy, lymphocytosis <strong>and</strong>occasional hepato-splenomegaly [133]. The diagnosisin typical cases is made clinically <strong>and</strong>/or serologically[69]. Histologically, <strong>the</strong> changes <strong>of</strong> a primary EBV infectioncan be dramatic [30]. The surface epi<strong>the</strong>lium<strong>of</strong> <strong>the</strong> palatine tonsils is <strong>of</strong>ten necrotic. Follicular hyperplasiawith fused or bizarre-shaped follicles <strong>and</strong>numerous tangible body macrophages <strong>and</strong> distendedinterfollicular areas with atypical immunoblasts, plasmacells, plasmacytoid lymphocytes <strong>and</strong> histiocytes,occasionally grouped around necrotic foci, are typical.Mitoses are numerous. Rare Reed-Sternberg-like cellswith single or multiple nuclei without nucleoli may bepresent beneath <strong>the</strong> crypt epi<strong>the</strong>lium. The <strong>of</strong>ten atypicalimmunoblasts may simulate lymphoma, but in infectiousmononucleosis, <strong>the</strong>y <strong>of</strong>ten merge with cells in<strong>the</strong> reactive follicles <strong>and</strong> paracortex (Fig. 6.6). The proliferatinglymphoid cells are predominantly activatedT lymphocytes with CD8-positive T-cells dominatingover CD4-positive T-cells. The immunoblasts can be <strong>of</strong>B-cell or T-cell type, <strong>and</strong> are occasionally CD30-positive,but CD15-negative [1]. Infected cells are reactivefor EBV nuclear antigen (EBNA) [2] <strong>and</strong> latent membraneprotein (LMP) [182, 186].The most important differential diagnosis <strong>of</strong> infectiousmononucleosis, especially in older patients, is <strong>the</strong>extranodal manifestation <strong>of</strong> Hodgkin’s lymphoma <strong>and</strong>non-Hodgkin’s lymphoma (large cell <strong>and</strong> immunoblastictypes). Immunoglobulin rearrangements <strong>and</strong> T-cellreceptor gene rearrangements are lacking in infectiousmononucleosis. The differential diagnosis <strong>of</strong> ulceratingtonsillitis includes infections with herpes virus hominis(usually Herpes simplex virus type 1). Herpes simplexvirus can produce a vesiculo-bullous pharyngitis<strong>and</strong> may involve <strong>the</strong> palatine tonsils [106, 195]. Rupture<strong>of</strong> <strong>the</strong> vesicles results in sharply circumscribed shallowulcers infiltrated by neutrophilic granulocytes. Infectedepi<strong>the</strong>lial cells show <strong>the</strong> characteristic multinucleatedgiant cells with nuclear viral inclusion. The lymphoidinfiltrate <strong>and</strong> hyperplasia <strong>of</strong> a Herpes simplex virus infectionmay mimic a NK/T-cell lymphoma [184]. GroupA Coxsackie’s virus also produces punched-out vesicles<strong>and</strong> is <strong>of</strong>ten associated with a concomitant Herpes simplexvirus infection. Rare systemic autoimmune diseases,like <strong>the</strong> anti-phospholipid antibody syndrome, maycause tonsillar ulcers [79].Chronic <strong>and</strong> recurrent tonsillitis are typically associatedwith respiratory syncytial virus, reactivation <strong>of</strong> latentEpstein-Barr virus, H. influenza <strong>and</strong> Staphylococcusaureus [44, 117, 204]. After episodes <strong>of</strong> recurrent tonsillitis,<strong>the</strong> palatine tonsils show extensive fibrosis at <strong>the</strong>site <strong>of</strong> <strong>the</strong> former capsule <strong>and</strong> scarring with entrapped,“pulled up” skeletal muscle fibres at <strong>the</strong> base <strong>of</strong> <strong>the</strong> tonsil<strong>and</strong> atrophic lymphoid tissue with small lymphoid follicleswith atrophic germinal centres. Granulomas maybe present. The crypts are distended <strong>and</strong> filled with keratinousdebris, inflammatory cells <strong>and</strong> occasional aggregates<strong>of</strong> actinomyces. Retention cysts can be formedwithin <strong>the</strong> deep crypts <strong>of</strong> chronically irritated palatinetonsils after occlusion <strong>of</strong> <strong>the</strong> orifice. The crypt epi<strong>the</strong>liumbecomes keratinised. Calcification <strong>of</strong> <strong>the</strong> desquamateddebris following deposition <strong>of</strong> inorganic saltsmay result in tonsillar calculi, so-called tonsilloliths. Thepresence <strong>of</strong> actinomyces is not associated causally withrecurrent tonsillitis [57, 120].Human immunodeficiency virus 1 (HIV-1) infectslymphocytes <strong>of</strong> lymph nodes <strong>and</strong> extranodal lymphoidtissue. Hypertrophy <strong>of</strong> <strong>the</strong> nasopharyngeal <strong>and</strong> palatinetonsils is among <strong>the</strong> earliest clinical manifestations <strong>of</strong>HIV-1 infections. Enlargement <strong>of</strong> <strong>the</strong> palatine tonsils isusually bilateral <strong>and</strong> large ulcers may lead to completedestruction <strong>of</strong> <strong>the</strong> tonsils [28]. The histological changesin HIV-induced tonsillar hypertrophy vary with stage<strong>and</strong> progression <strong>of</strong> <strong>the</strong> infection. The earliest changes includeflorid reactive follicular hyperplasia with irregularlyshaped germinal centres with an attenuated or absentmantle cell zone. Ano<strong>the</strong>r early change suggesting HIVinfection is “follicle lysis”, with permeation <strong>and</strong> disruption<strong>of</strong> germinal centres by “infiltrating” small lymphocytescreating a “moth eaten” appearance. Follicle lysis/follicular involution involves loss <strong>of</strong> tangible body macrophagesas well as <strong>the</strong> mantle zone <strong>of</strong> lymph follicles. Interfollicularhaemorrhage is ano<strong>the</strong>r feature <strong>of</strong> follicularinvolution. Multinucleated giant cells are a typical <strong>and</strong>specific feature <strong>of</strong> HIV tonsillitis. The multinucleated gi-


186 S. Regauer6abcdeFig. 6.6. EBV tonsillitis. a The hypertrophic palatine tonsil containsreactive lymph follicles which are distended by a dense interfollicularinfiltrate. b The surface epi<strong>the</strong>lium is also heavily infiltratedby numerous atypical lymphocytes, plasma cells, histiocytes.c–d The atypical large immunoblasts with prominent nucleolishow numerous mitoses <strong>and</strong> merge with <strong>the</strong> germinal centres.e The atypical immunoblasts may be binuclear atypical lymphoblastsant cells are arranged preferentially in clusters beneath<strong>the</strong> squamous surface <strong>and</strong> crypt epi<strong>the</strong>lium, <strong>and</strong> are onlyoccasionally seen in intrafollicular areas. They have afoamy cytoplasm <strong>and</strong> <strong>the</strong> nuclei are arranged at <strong>the</strong> periphery<strong>of</strong> <strong>the</strong> cells. The giant cells react with antibodiesto S-100 <strong>and</strong> CD68. O<strong>the</strong>r features <strong>of</strong> chronic HIV infectionare monocytoid B-cell hyperplasia, paracortical <strong>and</strong>interfollicular expansion with immunoblasts <strong>and</strong> plasmacells, <strong>of</strong>ten with Russell bodies, <strong>and</strong> prominent interfollicularclusters <strong>of</strong> high endo<strong>the</strong>lial venules. The histolog-


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 187ical features in patients with advanced disease are effacement<strong>of</strong> nodal architecture, loss <strong>of</strong> <strong>the</strong> normal lymphoidcell population with replacement by benign plasma cells<strong>and</strong> increased vascularity [53, 174, 197].Acute non-bacterial tonsillitis <strong>and</strong> hypertrophy canbe <strong>the</strong> first sign <strong>of</strong> a post-transplant lymphoproliferativedisorder [163]. In a study <strong>of</strong> 42 paediatric transplantpatients, 28% had involvement <strong>of</strong> Waldeyer’s ring [147].In immunosuppressed children in particular with rapidprogressive enlargement <strong>of</strong> <strong>the</strong> tonsils lymphoma shouldbe suspected [180].6.3.4 Benign Tumours<strong>of</strong> Waldeyer’s Ring6.3.4.1 Squamous PapillomaICD-O:8121/0Squamous papillomas represent <strong>the</strong> majority <strong>of</strong> benigntonsillar <strong>and</strong> oropharyngeal tumours [89]. They arise on<strong>the</strong> s<strong>of</strong>t palate <strong>and</strong> uvula, but also on <strong>the</strong> posterior wall<strong>of</strong> <strong>the</strong> oropharynx. They have an exophytic appearance.The fibrovascular stalk is covered in a regular, stratified,non-keratinising or keratinising squamous mucosa,with occasional parakeratosis. The vast majorityshow no viral cell changes. Some papillomas, however,show HPV-related cell changes with <strong>the</strong> typical koilocyteswith small pyknotic nuclei <strong>and</strong> a perinuclear halo,<strong>the</strong> so-called tonsillar condylomata [190]. Subtyping forHPV demonstrates typically low-risk HPV 6 <strong>and</strong> 11. Fora detailed description <strong>of</strong> squamous papilloma in <strong>the</strong> oropharynxsee Chap. 1.6.3.4.2 LymphangiomatousTonsillar PolypLymphangiomatous tonsillar polyps are benign tumours<strong>of</strong> <strong>the</strong> palatine tonsil, accounting for about 2%<strong>of</strong> all tonsillar neoplasms. They have been reportedby a number <strong>of</strong> different names such as angiomas,angi<strong>of</strong>ibromas, fibrolipoma, polypoid tumour containingfibro-adipose tissue <strong>and</strong> hamartomatous tonsillarpolyp <strong>and</strong> lymphangiectatic fibrous polyp [105]. Theyare pedunculated, mostly unilateral proliferations in<strong>the</strong> upper pole <strong>of</strong> <strong>the</strong> palatine tonsils in adults <strong>and</strong>children (age range <strong>of</strong> reported cases 3–63 years, witha median age <strong>of</strong> 26 years). Clinical symptoms are dysphagia,sore throat <strong>and</strong> <strong>the</strong> sensation <strong>of</strong> “a mass in <strong>the</strong>throat”. Lymphangiomatous tonsillar polyps measurebetween 0.5 <strong>and</strong> 4 cm. They are covered by respiratoryepi<strong>the</strong>lium or glycogenated or keratinised squamousepi<strong>the</strong>lium with foci <strong>of</strong> hyper- <strong>and</strong> parakeratosis.Clusters <strong>of</strong> lymphocytes are found within <strong>the</strong>squamous epi<strong>the</strong>lium (lymphocytic epi<strong>the</strong>liotropism)or in <strong>the</strong> submucosa beneath <strong>the</strong> basement membrane(Fig. 6.7). The stalk consists <strong>of</strong> dense fibroustissue, adipose tissues or myxoid <strong>and</strong> oedematousstroma, which contains numerous small to mediumsized,endo<strong>the</strong>lial-lined, lymphatic/vascular channelsfilled with proteinaceous fluid <strong>and</strong> lymphocytes[82]. Valves can be appreciated. Some endo<strong>the</strong>lialcells stain with antibodies to factor VIII, CD31 <strong>and</strong>CD34; o<strong>the</strong>rs are non reactive. The pathogenesis <strong>of</strong><strong>the</strong> lymphangiomatous polyps is uncertain. They maybe a hamartomatous proliferation, but <strong>the</strong>y may alsobe <strong>the</strong> result <strong>of</strong> a chronic inflammatory hyperplasia.Especially in children, lymphangiomatous polyps <strong>and</strong>papillary lymphoid polyps may be a manifestation <strong>of</strong>a chronic tonsillitis.6.3.5 Carcinomas<strong>of</strong> Waldeyer’s RingICD-O:8070/3Carcinomas <strong>of</strong> Waldeyer’s ring are typically squamouscell carcinomas (SCC) arising in <strong>the</strong> palatine tonsil <strong>and</strong><strong>the</strong> base <strong>of</strong> <strong>the</strong> tongue. They are more common in menthan in women <strong>and</strong> present during <strong>the</strong> 5th <strong>and</strong> 7th decades.Smoking, alcohol, poor hygiene, but also HPVinfections, are risk factors [32, 51, 160]. Some SCC maybe fungating <strong>and</strong> exophytic tumours, o<strong>the</strong>rs present asdeeply ulcerated infiltrative lesions. The majority <strong>of</strong>SCC <strong>of</strong> <strong>the</strong> palatine tonsil <strong>and</strong> base <strong>of</strong> <strong>the</strong> tongue typicallygrow undetected for some time as <strong>the</strong>y arise from<strong>the</strong> crypt epi<strong>the</strong>lium. At <strong>the</strong> time <strong>of</strong> clinical detection,extensive infiltration <strong>of</strong> <strong>the</strong> surrounding tissues <strong>and</strong>regional cervical lymph node metastases are typical.The metastases to cervical lymph nodes are <strong>of</strong>ten <strong>the</strong>presenting symptom <strong>of</strong> tonsillar carcinomas (Fig. 6.8).Histologically, primary carcinomas <strong>of</strong> <strong>the</strong> palatinetonsil <strong>and</strong> base <strong>of</strong> <strong>the</strong> tongue can be divided into keratinising<strong>and</strong> non-keratinising subtypes. The solidnon-keratinising carcinomas predominate. Tonsillarcarcinomas may show a “transitional type” differentiationresembling lymphoepi<strong>the</strong>lial carcinoma <strong>and</strong> EBVpositivity [111, 132]. A basaloid-squamous carcinoma<strong>of</strong> Waldeyer’s ring has also been described [7, 159]. Thelymph node metastases can be quite large <strong>and</strong> are <strong>of</strong>tencystic with multilocular complex lumina <strong>and</strong> papillaryprojections [165, 166, 189]. The majority <strong>of</strong> cysticmetastases are lined with a stratified epi<strong>the</strong>lium withcytological atypia <strong>and</strong> numerous mitoses. Foci <strong>of</strong> keratinisationcan be appreciated. The cysts mostly containnecrotic tumour cells <strong>and</strong> debris. A minority <strong>of</strong> <strong>the</strong>cystic lymph node metastases is filled with clear fluid.Fluid-filled cystic metastases are more common in carcinomas<strong>of</strong> <strong>the</strong> base <strong>of</strong> <strong>the</strong> tongue than those arisingfrom <strong>the</strong> palatine tonsils. It has been postulated that


188 S. Regauer6abcdeFig. 6.7. Lymphangiomatous tonsillar polyp. a Lymphangiomatouspolyps vary in size between several millimetres to severalcentimetres. The stalk may be composed <strong>of</strong> fibro-vascular stromaor adipose tissue. b The stroma may be fibrotic <strong>and</strong> contains numerousthin-walled vascular channels filled with proteinaceousffluid <strong>and</strong> lymphocytes. c–d The surface is covered by ei<strong>the</strong>r squamousepi<strong>the</strong>lium with numerous intraepi<strong>the</strong>lial lymphocytes orby respiratory epi<strong>the</strong>lium with a dense lymphocytic infiltrate. e–fMarkedly dilated lymphatic vessels accompanied by lymphocyticinfiltratecarcinomas that produce cystic metastases originatefrom <strong>the</strong> excretory ductal system <strong>of</strong> <strong>the</strong> submucosalminor salivary gl<strong>and</strong>s within <strong>the</strong> base <strong>of</strong> <strong>the</strong> tongue<strong>and</strong> <strong>the</strong> palatine tonsil [165].A search for <strong>the</strong> primary carcinoma within <strong>the</strong>pharynx in patients with clinically occult tumoursshould include multiple blind biopsies <strong>of</strong> <strong>the</strong> base <strong>of</strong><strong>the</strong> tongue <strong>and</strong> oro- <strong>and</strong> nasopharynx <strong>and</strong>/or ton-


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 189lymph node or carcinoma arising in a branchiogeniccyst is probably a hypo<strong>the</strong>tical entity. Reports <strong>of</strong> a supposedbranchiogenic carcinoma included an extremelywell-differentiated SCC arising in <strong>the</strong> background<strong>of</strong> longst<strong>and</strong>ing chronic inflammation <strong>and</strong> scarring,one carcinoma arising from pre-auricular ectodermalremnants <strong>of</strong> <strong>the</strong> first pharyngeal/branchial cleft <strong>and</strong>ano<strong>the</strong>r report <strong>of</strong> a well-differentiated mucoepidermoidbranchiogenic carcinoma [16, 154, 178]. Treatment<strong>of</strong> SCC <strong>of</strong> Waldeyer’s ring is surgical resectionwith a neck dissection.6.3.6 Malignant Lymphomas<strong>of</strong> Waldeyer’s ringabFig. 6.8. Tonsillar carcinoma. a Tonsillar squamous cell carcinomasfrequently arise from <strong>the</strong> crypt epi<strong>the</strong>lium. The carcinoma infiltratesdeeply into <strong>the</strong> surrounding structures including skeletalmuscles. b Cystic lymph node metastasis showing complex papillationswhich are lined by squamous epi<strong>the</strong>lium; <strong>the</strong> cyst contentis necrotic debrissillectomies. This practice has provided overwhelmingevidence that isolated carcinomas in neck lymphnodes are metastases <strong>and</strong> not so-called primary“branchiogenic carcinomas” within a cervical lymphnode. The evolution <strong>of</strong> in situ or invasive SCC fromnon-neoplastic squamous epi<strong>the</strong>lium through dysplasiais viewed as <strong>the</strong> most important criterion for <strong>the</strong>histopathological diagnosis <strong>of</strong> a primary branchiogeniccarcinoma. Primary cystic carcinoma arising in aThis section gives a brief overview <strong>of</strong> lymphomas <strong>of</strong>Waldeyer’s ring. For a more detailed description, includinggenetic characteristics <strong>of</strong> <strong>the</strong>se lymphomassee <strong>the</strong> WHO classification <strong>and</strong> <strong>the</strong> revised European-American classification <strong>of</strong> lymphoid neoplasms [74,95]. Extranodal lymphomas <strong>of</strong> Waldeyer’s ring constituteabout 5–10% <strong>of</strong> all lymphomas in <strong>the</strong> USA <strong>and</strong>Europe, about 15% in Hong Kong <strong>and</strong> about 10–20%in Japan. Of all lymphomas involving Waldeyer’s ring,80% are primary to this site <strong>and</strong> <strong>the</strong> tonsillar fossa is<strong>the</strong> most common location, followed by <strong>the</strong> nasopharynx<strong>and</strong> <strong>the</strong> base <strong>of</strong> <strong>the</strong> tongue. Up to 20% <strong>of</strong> patientswith tonsillar lymphoma have an associated gastrointestinalinvolvement. Clinical presentation is that <strong>of</strong>a localised neoplasm, sore throat, dysphagias, <strong>and</strong> incases <strong>of</strong> nasopharyngeal involvement cranial nerve,auditory <strong>and</strong> nasal symptoms. Between 85 <strong>and</strong> 90% <strong>of</strong>all non-Hodgkin’s lymphomas in Waldeyer’s ring are<strong>of</strong> <strong>the</strong> B-cell phenotype, <strong>the</strong> remainder are <strong>of</strong> <strong>the</strong> T-celltype, but regional differences have been reported [146,150]. The vast majority <strong>of</strong> Waldeyer’s ring lymphomasare high-grade lymphomas, with only less than 15%being <strong>of</strong> low grade [113]. The majority <strong>of</strong> AIDS-relatedextranodal head <strong>and</strong> neck lymphomas are aggressiveB-cell lymphomas <strong>of</strong> <strong>the</strong> Burkitt type or immunoblasticdiffuse large B-cell lymphomas [207].6.3.6.1 Mantle Cell LymphomaICD-O:9673/3Mantle cell lymphoma (or centrocytic (mantle cell) lymphomain <strong>the</strong> Kiel classification; diffuse, small cleavedcell type in <strong>the</strong> Working Formulation) constitutes about5% <strong>of</strong> all non-Hodgkin’s B-cell lymphomas. Commonextranodal sites are <strong>the</strong> spleen, bone marrow, gastrointestinaltract <strong>and</strong> Waldeyer’s ring, particularly <strong>the</strong> palatinetonsil.


190 S. Regauer66.3.6.2 Extranodal Marginal ZoneB-Cell Lymphoma<strong>of</strong> Mucosa-AssociatedLymphoid TissueICD-O:9699/3Mucosa-associated lymphoid tissue (MALT lymphomasor immunocytoma in <strong>the</strong> REAL classification; small lymphocytic,lymphoplasmacytoid, diffuse small cleaved celllymphoma in <strong>the</strong> Working Formulation) are low-gradeextranodal lymphomas that comprise about 8% <strong>of</strong> all B-cell lymphomas, typically in gastrointestinal locations.Up to 14% occur in <strong>the</strong> head <strong>and</strong> neck area, but MALTlymphomas arising in Waldeyer’s ring are exceptionallyrare [112, 145]. In epi<strong>the</strong>lial tissues, <strong>the</strong> MALT lymphomacells infiltrate <strong>the</strong> epi<strong>the</strong>lium, forming lymphoepi<strong>the</strong>liallesions. In Waldeyer’s ring, however, cautionis advised not to over-interpret <strong>the</strong> normally occurringintraepi<strong>the</strong>lial lymphocytes within <strong>the</strong> tonsillar <strong>and</strong>nasopharyngeal mucosa as lymphoma [94]. In generalMALT lymphomas run an indolent course with a tendencyto remain localised. They are sensitive to radiation<strong>the</strong>rapy. Patients have prolonged disease-free intervalsafter treatment, but recurrences may involve o<strong>the</strong>rextranodal sites.6.3.6.3 Extranodal NK/T-Cell Lymphoma, Nasal TypeICD-O:9719/3The extranodal NK/T-cell lymphoma, nasal type (orangiocentric T-cell lymphoma in <strong>the</strong> REAL classification;o<strong>the</strong>r historical names: lethal midline granuloma,malignant midline reticulosis, angiocentric immunoproliferativelesion) shows a predilection for <strong>the</strong>nasal cavity, nasopharynx <strong>and</strong> palate, but also occursin skin, s<strong>of</strong>t tissues, gastrointestinal tract <strong>and</strong> testis.The denominator “nasal type” indicates that <strong>the</strong> nasalcavity is <strong>the</strong> most common <strong>and</strong> prototypic site <strong>of</strong>involvement (Chap. 2). Some cases can be accompaniedby secondary lymph node involvement. Rapidsystemic dissemination is common, but bone marrowinvolvement is very rare. NK/T-cell lymphoma is morecommon in Asia, Mexico <strong>and</strong> South America than inEurope <strong>and</strong> North America, <strong>and</strong> shows a strong associationwith Epstein-Barr virus [24, 65, 99, 135, 142,148]. The prognosis <strong>of</strong> nasal NK/T-cell lymphoma isvariable despite aggressive <strong>the</strong>rapy. Some patients respondwell <strong>and</strong> o<strong>the</strong>rs die with disseminated disease.The prognostic influence <strong>of</strong> <strong>the</strong> cytological differentiationis unclear.6.3.6.4 Hodgkin’s LymphomaICD-O:9650/3Although exceptionally rare in extranodal sites, primaryHodgkin’s lymphoma does occur in Waldeyer’s ring<strong>and</strong> nasopharynx. In a review <strong>of</strong> 659 upper respiratorytract lymphomas, 6 cases <strong>of</strong> Hodgkin’s lymphoma inWaldeyer’s ring were identified [35, 103]. The WHO dividesHodgkin’s lymphomas into <strong>the</strong> nodular lymphocytepredominant Hodgkin’s lymphoma <strong>and</strong> <strong>the</strong> classicalHodgkin’s lymphoma (subdivided into lymphocyterich,nodular sclerosis, mixed cellularity <strong>and</strong> lymphocyte-depletedHodgkin’s lymphoma) [95]. Waldeyer’sring Hodgkin’s lymphomas belong predominantly to<strong>the</strong> classical subtypes: seven primary cases <strong>of</strong> nodularlymphocyte-rich Hodgkin lymphoma were reportedin two studies with a total <strong>of</strong> 27 patients [25, 179]. Inano<strong>the</strong>r series with 16 patients with Waldeyer’s ringHodgkin’s lymphoma, 50% were classified as mixedcellularity, 25% as nodular sclerosis, one case was anodular lymphocyte predominant subtype <strong>and</strong> threewere unclassified [103]. Epstein-Barr virus has beenpostulated to play a pathogenetic role in Hodgkin’slymphoma since it has been demonstrated in <strong>the</strong> majority<strong>of</strong> Hodgkin’s lymphomas <strong>of</strong> Waldeyer’s ring at ahigher incidence than in nodal Hodgkin’s lymphomas[81, 103]. In addition, patients <strong>of</strong>ten had a history <strong>of</strong>infectious mononucleosis. Therapy is local irradiationwith or without chemo<strong>the</strong>rapy.6.3.6.5 ExtramedullaryPlasmacytomaICD-O:9734/3The upper aerodigestive tract is <strong>the</strong> most common sitefor extra-osseous plasmacytomas, with 80% arising in<strong>the</strong> head <strong>and</strong> neck area. In a series <strong>of</strong> 299 cases, 22%arose in <strong>the</strong> nasopharynx <strong>and</strong> 7% in <strong>the</strong> tonsil [55, 102,124]. Plasmacytomas <strong>of</strong> Waldeyer’s ring proceed less<strong>of</strong>ten to multiple myeloma than those <strong>of</strong> o<strong>the</strong>r locations.6.3.7 Systemic Disease AffectingWaldeyer’s RingTangier’s disease (familial hypo-alpha-lipoproteinemia)is an autosomal-recessive metabolic disease with a deficiency<strong>of</strong> high-density lipoproteins <strong>and</strong> extremely lowlevels <strong>of</strong> plasma cholesterol [85]. The deranged fat metabolismresults in storage <strong>of</strong> cholesterol esters in <strong>the</strong>reticuloendo<strong>the</strong>lial system <strong>and</strong> macrophages <strong>of</strong> <strong>the</strong> pharyngeal<strong>and</strong> gastrointestinal tract mucosa, but also insmooth muscle cells, pericytes <strong>and</strong> Schwann cells <strong>of</strong> pe-


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 191ripheral nerves. Clinically, most patients are asymptomatic,but children with Tangier’s disease have enlarged,hyperplastic palatine <strong>and</strong> pharyngeal tonsils with yellow-orangeor yellow-grey discoloration. Histologically,large groups <strong>and</strong> accumulations <strong>of</strong> macrophages withfoamy cytoplasm can be identified in <strong>the</strong> palatine <strong>and</strong>pharyngeal tonsils [6, 48].A m y l o i d o s i s is usually a systemic disease <strong>of</strong> multifactorialorigin that may involve <strong>the</strong> head <strong>and</strong> neckarea. Particularly <strong>the</strong> upper respiratory tract is commonlyaffected by amyloidosis with a symmetrical enlargement<strong>of</strong> <strong>the</strong> tongue. Small amounts <strong>of</strong> amyloid depositionin Waldeyer’s ring have been described in plasmacytomas,nasopharyngeal carcinomas or tonsillitis.Isolated tumour-like involvement <strong>of</strong> <strong>the</strong> nasopharynxwith <strong>and</strong> without immunoglobulin light chain restriction,<strong>of</strong> <strong>the</strong> entire Waldeyer’s ring or <strong>the</strong> palatine tonsilswithout systemic disease is exceptionally rare [11,45, 116, 156].Involvement <strong>of</strong> <strong>the</strong> naso- <strong>and</strong> oropharynx by systemicsarcoidosis is well documented [127]. Unsuspectedisolated sarcoidosis <strong>of</strong> <strong>the</strong> palatine <strong>and</strong> pharyngealtonsils in <strong>the</strong> absence <strong>of</strong> systemic disease is veryrare [46, 126]. Histologically, <strong>the</strong> sarcoidosis granulomasare composed <strong>of</strong> densely packed epi<strong>the</strong>lioid histiocytes<strong>and</strong> macrophages without central necrosis.Differential diagnoses include recurrent tonsillitis,which may feature giant cells <strong>and</strong> even foreign bodygiant cell granulomas. Ano<strong>the</strong>r differential diagnosisconsists <strong>of</strong> infections with mycobacterium tuberculosis<strong>and</strong> formation <strong>of</strong> caseating tuberculoid granulomas.The majority <strong>of</strong> patients with pulmonary tuberculosishave nasopharyngeal involvement, but isolatednasopharyngeal tuberculosis is rare [2, 161, 193].Metastases from primary tumours outside <strong>the</strong> head<strong>and</strong> neck area to <strong>the</strong> naso- <strong>and</strong> oropharynx are exceptionallyrare, because <strong>the</strong> nasopharyngeal <strong>and</strong> palatinetonsils have no afferent lymph vessels. Consequently,most metastatic tumours in tonsils represent haematogenousdeposits. Bilateral metastases to <strong>the</strong> palatinetonsils have been described for pancreatic carcinoma[131]. O<strong>the</strong>r metastatic tumours to <strong>the</strong> palatine tonsilinclude carcinomas <strong>of</strong> <strong>the</strong> breast, lung, stomach, colon,prostate, skin <strong>and</strong> kidneys [173]. Renal cell carcinomasare known to metastasise to <strong>the</strong> nasopharynx[169].References1. Abbondanzo S, Sato N, Straus SE, Jaffe ES (1990) Acute infectiousmononucleosis. Am J Clin Pathol 93:698–7022. Aktan B, Selimoglu E, Ucuncu H, Sutbeyaz Y (2002) Primarynasopharyngeal tuberculosis in a patient with <strong>the</strong> complaint<strong>of</strong> snoring. J Laryngol Otol 116:301–3033. Albers FW, Cuvelier CA, Renders MT, Cauwenberge PB(1993) Oncocytoma <strong>of</strong> <strong>the</strong> nasopharynx. Rhinol 31:41–434. Al-Sarraf M, Reddy MS (2002) Nasopharyngeal carcinoma.Curr Treat Options Oncol 3:21–235. Au WY, Kwong YL, Ho WK, Shek TW (2001) Primary granulocyticsarcoma <strong>of</strong> <strong>the</strong> nasopharynx. Am J Hematol 67:273–2746. Bale P, Clifton-Bligh P, Benjamin B, Whyte H (1971) <strong>Pathology</strong><strong>of</strong> Tangier disease. J Clin Pathol 24:609–6167. Banks ER, Frierson HF Jr, Mills SE, George E, Zarbo RJ,Swanson PE (1992) Basaloid squamous cell carcinoma <strong>of</strong> <strong>the</strong>head <strong>and</strong> neck. A clinicopathologic <strong>and</strong> immunohistochemicalstudy <strong>of</strong> 40 cases. Am J Surg Pathol 16:939–9468. Batsakis JG, Solomon AR, Rice DH (1981) The pathology <strong>of</strong>head <strong>and</strong> neck tumors: carcinoma <strong>of</strong> <strong>the</strong> nasopharynx, part11. <strong>Head</strong> <strong>Neck</strong> Surg 3:511–5249. Beham A, Beham-Schmid C, Regauer S, Auböck L, StammbergerH (2000) Nasopharyngeal angi<strong>of</strong>ibroma: true neoplasmor vascular malformation? Adv Anat Pathol 7:36–4610. Beham-Schmid C, Beham A, Jakse R, Auböck L, Höfler G(1998) Extranodal follicular dendritic cell tumour <strong>of</strong> <strong>the</strong> nasopharynx.Virchows Arch 432:293–29811. Beiser M, Messer G, Samuel J, Gross B, Shanon E (1980) Amyloidosis<strong>of</strong> Waldeyer’s ring. A clinical <strong>and</strong> ultrastructuralreport. Acta Otolaryngol 89:562–56912. Benke TT, Zitsch RP, Nashelsky MB (1995) Bilateral oncocyticcysts <strong>of</strong> <strong>the</strong> nasopharynx. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg112:321–32413. Boccon-Gibod LA, Grangeponte MC, Boucheron S, JossetPP, Roger G, Berthier-Falissard ML (1996) Salivary gl<strong>and</strong> anlagetumor <strong>of</strong> <strong>the</strong> nasopharynx: a clinicopathologic <strong>and</strong> immunohistochemicalstudy <strong>of</strong> three cases. Pediatr Pathol LabMed 16:973–98314. Bossen EH, Hudson WR (1987) Oligodendroglioma arisingin heterotopic brain tissue <strong>of</strong> <strong>the</strong> s<strong>of</strong>t palate <strong>and</strong> nasopharynx.Am J Surg Pathol 11:571–57415. Bremer JW, Neel HB, DeSanto LW, Jones GC (1986) Angi<strong>of</strong>ibroma:treatment trends in 150 patients during 40 years Laryngoscope96:1321–132916. Browder JP, Wheeler MS, Henley JT, Geratz JD (1984) Mucoepidermoidcarcinoma in a cervical cyst: a case <strong>of</strong> branchiogeniccarcinoma? Laryngoscope 94:107–11217. Burns BV, Axon PR, Pahade A (2001) ‘Hairy polyp’ <strong>of</strong> <strong>the</strong>pharynx in association with an ipsilateral branchial sinus:evidence that <strong>the</strong> ‘hairy polyp’ is a second branchial archmalformation. J Laryngol Otol 115:145–14818. Byrne MN, Session DG (1990) Nasopharyngeal craniopharyngioma.Case report <strong>and</strong> literature review. Ann Otol RhinolLaryngol 99:633–63919. Campbell WM, McDonald TJ, Unni KK (1980) Nasal <strong>and</strong> paranasalpresentations <strong>of</strong> chordomas. Laryngoscope 90:612–61820. Canalis RF, Jenkins HA, Hemenway WG, Lincoln C (1978)Nasopharyngeal rhabdomyosarcoma. A clinical perspective.Arch Otolaryngol 104:122–12621. Carrau RL, Petruzzelli G, Cass SP (1995) Adenoid cystic carcinoma<strong>of</strong> <strong>the</strong> nasopharynx. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg112:501–50222. Chan AC, Chan KW, Chan JK, Au WY, Ho WK, Ng WM(2001) Development <strong>of</strong> follicular dendritic cell sarcoma inhyaline-vascular Castleman’s disease <strong>of</strong> <strong>the</strong> nasopharynx:tracing its evolution by sequential biopsies. Histopathology38:510–51823. Chan JK, Tsang WY, Ng CS (1994) Follicular dendritic celltumor <strong>and</strong> vascular neoplasm complicating hyaline-vascularCastleman’s disease. Am J Surg Pathol 18:517–525


192 S. Regauer624. Chan JK, Yip TT, Tsang WY, Ng CS, Lau WH, Poon YF, WongCC, Ma VW (1994) Detection <strong>of</strong> Epstein-Barr viral RNA inmalignant lymphomas <strong>of</strong> <strong>the</strong> upper aerodigestive tract. Am JSurg Pathol 18:938–94625. Chang KL, Kamel OW, Arber DA, Horyd ID, Weiss LM(1995) Pathologic features <strong>of</strong> nodular lymphocyte predominanceHodgkin’s disease in extranodal sites. Am J SurgPathol 19:1313–132426. Chao TY, Chow KC, Chang JY, Wang CC, Tsao TY, Harn HJ,Chi KH (1996) Expression <strong>of</strong> Epstein-Barr virus-encodedRNAs as a marker for metastatic undifferentiated nasopharyngealcarcinoma. Cancer 78:24–2927. Chaudhry AP, Lore JM Jr, Fisher JE, Gambrino AG (1978)So-called hairy polyps or teratoid tumors <strong>of</strong> <strong>the</strong> nasopharynx.Arch Otolaryngol 104:517–52528. Chaudhry R, Akhtar S, Lucente FE, Kim DS (1996) Largeoral ulcers leading to destruction <strong>of</strong> <strong>the</strong> tonsils in patientswith AIDS. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 114:474–47829. Chessin H, Urdaneta N, Smith H, Gilder JV (1976) Chromophobeadenoma manifesting as a nasopharyngeal mass. ArchOtolaryngol 102:631–63330. Childs C, Parham DM, Berard CW (1987) Infectious mononucleosis.The spectrum <strong>of</strong> morphologic changes simulatinglymphoma in lymph nodes <strong>and</strong> tonsils. Am J Surg Pathol11:122–13231. Choi P, Suen MW, Huang DP, Lo KW, Lee JC (1970) Nasopharyngealcarcinoma: genetic changes, Epstein-Barr virusinfection, or both. A clinical <strong>and</strong> molecular study <strong>of</strong> 36 patients.Cancer 72:2873–287832. Civantos FJ, Goodwin WJJ (1996) Cancer <strong>of</strong> <strong>the</strong> oropharynx.In: Myers EN, Suen YJ (1996) Cancer <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck.Saunders, Philadelphia, pp 381–38033. Civen R, Vaisanen ML, Finegold SM (1993) Peritonsillar abscess,retropharyngeal abscess, mediastinitis, <strong>and</strong> nonclostridialanaerobic myonecrosis: a case report. Clin Infect Dis16:299–30334. C<strong>of</strong>fin C, Swanson P, Wick M, Dehner L (1993) Chordoma inchildhood <strong>and</strong> adolescence. A clinicopathologic analysis <strong>of</strong>12 cases. Arch Pathol Lab Med 117:927–93335. Colby TV, Hoppe RT, Warnke RA (1981) Hodgkin’s disease:a clinicopathologic study <strong>of</strong> 659 cases. Cancer 49:1848–185836. Coppit GL III, Perkins JA, Manning SC (2000) Nasopharyngealteratomas <strong>and</strong> dermoids: a review <strong>of</strong> <strong>the</strong> literature <strong>and</strong>case series. Int J Pediatr Otorhinolaryngol 52:219–22737. Cummings BJ, Blend R, Fitzpatrick P, Clark R, Harwood A,Keane T, Beale F, Garrett P, Payne D, Rider W (1984) Primaryradiation <strong>the</strong>rapy for juvenile nasopharyngeal angi<strong>of</strong>ibroma.Laryngoscope 94:1599–160538. Dehner LP, Valbuena L, Perez-Atayde A, Reddick RL, AskinFB, Rosai J (1994) Salivary gl<strong>and</strong> anlage tumor (“congenitalpleomorphic adenoma”). A clinicopathologic, immunohistochemical<strong>and</strong> ultrastructural study <strong>of</strong> nine cases. Am JSurg Pathol 18:25–2639. Deutsch M, Orl<strong>and</strong>o SJ (1999) Re-irradiation for recurrentrhabdomyosarcoma <strong>of</strong> <strong>the</strong> nasopharynx in a child. Oral Oncol35:343–34540. Dictor M, Siven M, Tennvall J, Rambech E (1995) Determination<strong>of</strong> non-endemic nasopharyngeal carcinoma by in situhybridization for Epstein-Barr virus EBER1 RNA: sensitivity<strong>and</strong> specificity in cervical node metastases. Laryngoscope105:407–41241. Dolen WK, Sp<strong>of</strong>ford B, Selner JC (1990) The hidden tonsils <strong>of</strong>Waldeyer’s ring. Ann Allergy 65:244–24842. Dubey SP, Sengupta SK, Vele DD (1996) Nasopharyngeal osteosarcomaas second malignant neoplasm in a post-treatedunilateral retinoblastoma: report <strong>of</strong> a case <strong>and</strong> review <strong>of</strong> literature.Int J Pediatr Otorhinolaryngol 34:265–27143. Easton J, Levine PH, Hyams VJ (1980) Nasopharyngeal carcinomain <strong>the</strong> United States. A pathologic study <strong>of</strong> 177 US<strong>and</strong> 30 foreign cases. Arch Otolaryngol 106:88–9144. Endo LH, Ferreira D, Montenegro MCS, Pinto GA, AltemaniA, Bortoleto AE Jr, Vassallo J (2001) Detection <strong>of</strong> Epstein-Barr virus in tonsillar tissue <strong>of</strong> children <strong>and</strong> <strong>the</strong> relationshipwith recurrent tonsillitis. Int J Pediatr Otorhinolaryngol58:9–1545. Eriksen HE (1970) A case <strong>of</strong> primary localized amyloidosis inboth tonsils. J Laryngol Otol 84:525–53146. Erwin SA (1989) Unsuspected sarcoidosis <strong>of</strong> <strong>the</strong> tonsil. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 100:245–24747. Ferouz AS, Mohr RM, Paul P (1995) Juvenile nasopharyngealangi<strong>of</strong>ibroma <strong>and</strong> familial adenomatous polyposis: an association?Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 113:435–43948. Ferrans V, Fredrickson D (1975) The pathology <strong>of</strong> Tangierdisease. A light <strong>and</strong> electron microscopic study. Am J Pathol78:101–15849. Fletcher CDM, Unni KK, Mertens F (2002) World Health OrganizationClassification <strong>of</strong> Tumours. In: Kleihues P, SobinLH (eds) <strong>Pathology</strong> <strong>and</strong> genetics <strong>of</strong> tumors <strong>of</strong> s<strong>of</strong>t tissue <strong>and</strong>bone. IARC, Lyon50. Folpe AL, McKenney JK, Bridge JA, Weiss SW (2002) Sclerosingrhabdomyosarcoma in adults: report <strong>of</strong> four cases <strong>of</strong> ahyalinizing, matrix-rich variant <strong>of</strong> rhabdomyosarcoma thatmay be confused with osteosarcoma, chondrosarcoma, orangiosarcoma. Am J Surg Pathol 26:1175–118351. Fouret P, Monceaux G, Temam S, Lacourreye L, Guily JLS(1997) Human papillomavirus in head <strong>and</strong> neck squamouscell carcinomas in nonsmokers. Arch Otolaryngol <strong>Head</strong><strong>Neck</strong> Surg 123:513–51652. Franco V, Florena AM, Lombardo F, Restivo S (1996) Bilateralhairy polyp <strong>of</strong> <strong>the</strong> oropharynx. J Laryngol Otol 110:288–29053. Frankel S, Wenig B, Ferlito A (1997) Human immunodeficiencyvirus-1 infection <strong>of</strong> <strong>the</strong> lymphoid tissues <strong>of</strong> Waldeyer’sring. Ann Otol Rhinol Laryngol 106:611–61854. Fu YS, Perzin KH (1974) Non-epi<strong>the</strong>lial tumors <strong>of</strong> <strong>the</strong> nasalcavity, paranasal sinuses <strong>and</strong> nasopharynx: a clinicopathologicstudy. III. Cartilaginous tumors (chondroma, chondrosarcoma).Cancer 34:453–46355. Fu YS, Perzin KH (1978) Nonepi<strong>the</strong>lial tumors <strong>of</strong> <strong>the</strong> nasalcavity, paranasal sinuses <strong>and</strong> nasopharynx. A clinicopathologicstudy. IX. Plasmacytoma. Cancer 42:2399–240656. Gadwal SR, Fanburg-Smith JC, Gannon FH, Thompson LD(2000) Primary chondrosarcoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck in pediatricpatients: a clinicopathologic study <strong>of</strong> 14 cases with areview <strong>of</strong> <strong>the</strong> literature. Cancer 88:2181–218857. Gaffney R, Harrison M, Walsh M, Sweeney E, Cafferkey M(1993) The incidence <strong>and</strong> role <strong>of</strong> actinomyces in recurrentacute tonsillitis. Clin Otolaryngol 18:268–27158. Gallo O, Bianchi S, Giannini A, Gallina E, Libonati G, Fini-Storchi O (1991) Correlation between histopathological <strong>and</strong>biological findings in nasopharyngeal carcinoma <strong>and</strong> itsprognostic significance. Laryngoscope 101:487–59359. Gaudecker B von (1988) Development <strong>and</strong> functional anatomy<strong>of</strong> <strong>the</strong> human tonsilla palatina. Acta Otolaryngol Suppl454:28–3260. Gaudecker B von, Muller-Hermelink H (1982) The development<strong>of</strong> <strong>the</strong> human tonsilla palatina. Cell Tissue Res 224:579–60061. Gerber JE (2001) Acute necrotizing bacterial tonsillitis withClostridium perfringens. Am J Forensic Med Pathol 22:177–17962. Giardiello FM, Hamilton SR, Krush AJ, Offerhaus JA, BookerSV, Petersen GM (1993) Nasopharyngeal angi<strong>of</strong>ibroma inpatients with familial adenomatous polyposis. Gastroenterology105:1550–155263. Giffler RF, Gillespie JJ, Ayala AG, Newl<strong>and</strong> JR (1977) Lymphoepi<strong>the</strong>liomain cervical lymph nodes <strong>of</strong> children <strong>and</strong>young adults. Am J Surg Pathol 1:293–30264. Goldenberg D, Golz A, Netzer A, Flax-Goldenberg R,Joachims HZ (2001) Synergistic necrotizing cellulitis as acomplication <strong>of</strong> peritonsillar abscess. Am J Otolaryngol22:415–41965. Gorp J van, Weiping L, Jacobse K, Liu YH, Li FY, Weger RAde, Li G (1994) Epstein-Barr virus in nasal T-cell lymphomas(polymorphic reticulosis/midline malignant reticulosis)in western China. J Pathol 173:81–87


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 19366. Graeme-Cook F, Bhan AK, Harris NL (1993) Immunohistochemicalcharacterization <strong>of</strong> intraepi<strong>the</strong>lial <strong>and</strong> subepi<strong>the</strong>lialmononuclear cells <strong>of</strong> <strong>the</strong> upper airways. Am J Pathol143:1416–142267. Graziani N, Donnet A, Bugha TN, Dufour H, Figarella-Branger D, Grisoli F (1994) Ectopic basisphenoidal craniopharyngioma:case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Neurosurgery34:346–34968. Grewal D, Hiran<strong>and</strong>ani N, Kalgutkar J (1985) Congenital absence<strong>of</strong> <strong>the</strong> palatine tonsil associated with congenital malformation<strong>of</strong> <strong>the</strong> external ear. A congenital anomaly. J LaryngolOtol 99:285–28869. Gulley ML, Amin MB, Nicholls JM, Banks PM, Ayala AG,Srigley JR, Eagan PA, Ro JY (1995) Epstein-Barr virus is detectedin undifferentiated nasopharyngeal carcinoma but notin lymphoepi<strong>the</strong>lioma-like carcinoma <strong>of</strong> <strong>the</strong> urinary bladder.Hum Pathol 26:1207–121470. Gürtl B, Beham A, Zechner R, Stammberger H, Höfler G(2000) Nasopharyngeal angi<strong>of</strong>ibroma: an APC-gene-associatedtumor? Hum Pathol 31:1411–141371. Haddad Jr J, Senders CW, Leach CS, Stool SE (1990) Congenitalhairy polyp <strong>of</strong> <strong>the</strong> nasopharynx associated with cleftpalate: report <strong>of</strong> two cases. Int J Pediatr Otorhinolaryngol20:127–13572. Hama S, Arita K, Nishisaka T, Fukuhara T, Tominaga A, SugiyamaK, Yoshioka H, Eguchi K, Sumida M, Heike Y, KurisuK (2002) Changes in <strong>the</strong> epi<strong>the</strong>lium <strong>of</strong> Rathke cleft cyst associatedwith inflammation. J Neurosurg 96:209–21673. Har-El G, Zirkin H, Tovi F, Sidi J (1985) Congenital pleomorphicadenoma <strong>of</strong> <strong>the</strong> nasopharynx (report <strong>of</strong> a case). J Otolaryngol99:25–3674. Harris NL, Jaffe ES, Stein H, Banks PM, Chan JKC, ClearyML, Delsol G, Wolf-Peeters CD, Falini B, Gatter KC, GroganTM, Isaacson PG, Knowles DM, Mason DY, Muller-HermelinkHK, Pileri SA, Piris MA, Ralfkiaer E, Warnke RA (1994)A revised European-American classification <strong>of</strong> lymhoid neoplasms:a proposal from <strong>the</strong> International Lymphoma StudyGroup. Blood 84:1361–139275. Harrison DFN (1987) The natural history, pathogenesis, <strong>and</strong>treatment <strong>of</strong> juvenile angi<strong>of</strong>ibroma. Arch Otolaryngol <strong>Head</strong><strong>Neck</strong> Surg 113:936–94276. Hasselt CA van, Ng NK (1991) Papillary adenocarcinoma <strong>of</strong><strong>the</strong> nasopharynx. J Laryngol Otol 105:853–85477. Hawkins E, Krischer JP, Smith BE, Hawkins HK, FinegoldMJ (1990) Nasopharyngeal carcinoma in children – a retrospectivereview <strong>and</strong> demonstration <strong>of</strong> Epstein-Barr viral genomesin tumor cell cytoplasm. A report <strong>of</strong> <strong>the</strong> Pediatric OncologyGroup. Hum Pathol 21:805–81078. Heffner DK, Thompson LD, Schall DG, Anderson V (1996)Pharyngeal dermoids (“hairy polyps”) as accessory auricles.Ann Otol Rhinol Laryngol 105:819–82479. Henning B, Buchner N, Kirchner J, Gillessen A (2000) Antiphospholipidantibody syndrome as <strong>the</strong> cause <strong>of</strong> clinicalrapidly progressing vasculopathy. Dtsch Med Wochenschr125:589–59380. Herait P, Ganem G, Lipinski M, Carlu C, Micheau C, SchwaabG, The G, Tursz T (1987) Lymphocyte subsets in tumour <strong>of</strong>patients with undifferentiated nasopharyngeal carcinoma:presence <strong>of</strong> lymphocytes with <strong>the</strong> phenotype <strong>of</strong> activated T-cells. Br J Cancer 55:135–13981. Herbst H, Niedobitek G, Kneba M (1990) High incidence<strong>of</strong> Epstein-Barr virus genomes in Hodgkin’s disease. Am JPathol 137:13–1882. Hiraide F, Inouye T, Tanaka E (1985) Lymphangiectatic fibrouspolyp <strong>of</strong> <strong>the</strong> palatine tonsil. A report <strong>of</strong> three cases. JLaryngol Otol 99:403–40983. Hirayama T (1978) Descriptive <strong>and</strong> analytical epidemiology<strong>of</strong> nasopharyngeal cancer. Ablashi DV, De-The G, Ito ?Y (eds)Nasopharyngeal carcinoma: etiology <strong>and</strong> control. IARC,Lyon, pp 167–18984. Hjertaas RJ, Morrison MD, Murray RB (1979) Teratomas <strong>of</strong><strong>the</strong> nasopharynx. J Otolaryngol 8:411–41685. Hobbs HH, Rader DJ (1999) ABC1: connecting yellow tonsils,neuropathy, <strong>and</strong> very low HDL. J Clin Invest 104:1015–101786. Hollender A (1946) The nasopharynx. A study <strong>of</strong> 140 autopsyspecimens. Laryngoscope 56:282–30487. Hollowood K, Pease C, Mackay AM, Fletcher CDM (1991)Sarcomatoid tumours <strong>of</strong> lymph nodes showing folliculardendritic cell differentiation. J Pathol 103:90–9788. Hsiao J, Jin Y, Tsai S (1998) EBER1 in situ hybridization as anadjuvant for diagnosis <strong>of</strong> recurrent nasopharyngeal carcinoma.Anticancer Res 18:4585–458989. Hyams VJ (1978) Differential diagnosis <strong>of</strong> neoplasia <strong>of</strong> <strong>the</strong>palatine tonsil. Clin Otolaryngol 3:117–12690. Ikeda H, Yoshimoto T (2002) Clinicopathological study <strong>of</strong>Rathke’s cleft cysts. Clin Neuropathol 21:82–9191. Ikram M, Khan MAA, Ahmed M, Siddiqui T, Mian MY(2000) The histopathology <strong>of</strong> routine tonsillectomy specimens:results <strong>of</strong> a study <strong>and</strong> review <strong>of</strong> literature. Ear NoseThroat J 79:880–88292. Ikushima I, Korogi Y, Makita O, Komohara Y, Kawano H,Yamura M, Arikawa K, Takahashi M (1999) MR imaging <strong>of</strong>Tornwaldt’s cysts. AJR Am J Roentgenol 172:1663–166593. Inoue H, Sato Y, Tsuchiya B, Nagai H, Takahashi H, KameyaT (2002) Expression <strong>of</strong> Epstein-Barr virus-encoded smallnuclear RNA 1 in Japanese nasopharyngeal carcinoma. ActaOtolaryngol Suppl (547)113–11794. Jaffe ES (2002) Lymphoid lesions <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck: amodel <strong>of</strong> lymphocyte homing <strong>and</strong> lymphomagenesis. ModPathol 15:255–26395. Jaffe ES, Harris NL, H. HS, Vardiman VW (2001) <strong>Pathology</strong><strong>and</strong> genetics <strong>of</strong> tumors <strong>of</strong> haematopoietic <strong>and</strong> lymphoid tissues.In: Kleihues P, Sobin LH (eds) WHO Classification <strong>of</strong>Tumours. IARC, Lyon96. Job A, Walter N, David TM (1991) Solitary fibrous tumor <strong>of</strong><strong>the</strong> nasopharynx. J Laryngol Otol 105:213–21497. Jousimies-Somer H, Savolainen S, Makitie A, Ylikoski J(1993) Bacteriologic findings in peritonsillar abscesses inyoung adults. Clin Infect Dis 16:292–29898. Kamino H, Huang SJ, Fu YS (1988) Keratin <strong>and</strong> involucrinimmunohistochemistry <strong>of</strong> nasopharyngeal carcinoma. Cancer61:1142–114899. Kanavaros P, Lescs MC, Briere J, Divine M, Galateau F, JoabI, Bosg J, Farcet JP, Reyes F, Gaulard P (1993) Nasal T-celllymphoma: a clinicopathologic entity associated with peculiarphenotype <strong>and</strong> with Epstein-Barr virus. Blood 81:2688–2695100. Kanegane H, Kanegane C, Yachie A, Miyawaki T, Tosato G(1997) Infectious mononucleosis as a disease <strong>of</strong> early childhoodin Japan caused by primary Epstein-Barr virus infection.Acta Paediatr Jpn 39:166–171101. Kanoh N, Nishimura Y, Nakamura M, Mori M, Uematsu K(1991) Primary nasopharyngeal paraganglioma: a case report.Auris Nasus Larynx 18:307–314102. Kapadia SB, Desai U, Cheng VS (1982) Extramedullary plasmacytoma<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. A clinicopathologic study <strong>of</strong>20 cases. Medicine 61:317–329103. Kapadia SB, Roman LN, Kingma DW, Jaffe ES, Frizzera G(1995) Hodgkin’s disease <strong>of</strong> Waldeyer’s ring. Clinical <strong>and</strong>histoimmunophenotypic findings <strong>and</strong> association with Epstein-Barrvirus in 16 cases. Am J Surg Pathol 19:1431–1439104. Karajannis MA, Hummel M, Anagnostopoulos I, Stein H(1997) Strict lymphotropism <strong>of</strong> Epstein-Barr virus duringacute infectious mononucleosis in nonimmunocompromisedindividuals. Blood 89:2856–2862105. Kardon DE, Wenig BM, Heffner DK, Thompson LD (2000)Tonsillar lymphangiomatous polyps: a clinicopathologic series<strong>of</strong> 26 cases. Mod Pathol 13:1128–1133106. Karnauchow PN, Kaul WH (1988) Chronic herpetic laryngitiswith oropharyngitis. Ann Otol Rhinol Laryngol 97:286–288107. Kaur A, Shetty SC, Prasad D, Nirmala V (1997) Primary ectopicmeningioma <strong>of</strong> <strong>the</strong> palatine tonsil – a case report. J LaryngolOtol 111:179–181108. Kawashiro N, Koga K, Tsuchihashi N, Araki A (1994) Choanalatresia <strong>and</strong> congenital pharyngeal stenosis. Acta Otolaryngol517:27–32


194 S. Regauer6109. Kieff DA, Curtin HD, Limb CJ, Nadol JB (1998) A hairy polyppresenting as a middle ear mass in a pediatric patient. AmJ Otolaryngol 19:228–231110. Kikuchi K, Kowada M, Sasaki J, Sageshima M (1994) Largepituitary adenoma <strong>of</strong> <strong>the</strong> sphenoid sinus <strong>and</strong> <strong>the</strong> nasopharynx:report <strong>of</strong> a case with ultrastructural evaluations. SurgNeurol 42:330–334111. Klijanienko J, Micheau C, Azli N (1989) Undifferentiatedcarcinoma <strong>of</strong> nasopharyngeal type <strong>of</strong> tonsil. Arch Otolaryngol(<strong>Head</strong> <strong>Neck</strong> Surg) 115:731–734112. Kojima M, Tamaki Y, Nakamura S (1993) Malignant lymphoma<strong>of</strong> Waldeyer’s ring. A histological <strong>and</strong> immunohistochemicalstudy. APMIS 101:537–544113. Krol ADG, Cessie SL, Snijder S, Kluin-Nelemans JC, KluinPM, Noordijk EM (2001) Waldeyer’s ring lymphomas: a clinicalstudy from <strong>the</strong> Comprehensive Cancer Center West populationbased NHL registry. Leuk Lymphoma 42:1005–1013114. Kuo T, Tsang NM (2001) Salivary gl<strong>and</strong> type nasopharyngealcarcinoma: a histologic, immunohistochemical, <strong>and</strong> Epstein-Barrvirus study <strong>of</strong> 15 cases including a psammomatousmucoepidermoid carcinoma. Am J Surg Pathol 25:80–86115. Lee AW, Poon YP, Foo W, Law SC, Cheung FK, Chan DK,Tung SY, Thaw M, Ho JH. (1992) Retrospective analysis <strong>of</strong>5037 patients with nasopharyngeal cancer treated during1976-1985: overall survival <strong>and</strong> patterns <strong>of</strong> failure. Int J RadiolOncol Biol Phys 23:261–270116. Lim JS, Lebowitz RA, Jacobs JB (1999) Primary amyloidosispresenting as a nasopharyngeal mass. Am J Rhinol 13:209–212117. Lindroos R (2000) Bacteriology <strong>of</strong> <strong>the</strong> tonsil core in recurrenttonsillitis <strong>and</strong> tonsillar hypertrophy – a short review. ActaOtolaryngol Suppl 543:206–208118. Lloyd G, Howard D, Lund VJ, Savy L (2000) Imaging for juvenileangi<strong>of</strong>ibroma. J Laryngol Otol 114:727–730119. MacDonald MR, Freeman JL, Hui MF, Cheung RK, WardeP, Mclvor NP, Irish J, Dosch HM (1995) Role <strong>of</strong> Epstein-Barrvirus in fine-needle aspirates <strong>of</strong> metastatic neck nodes in <strong>the</strong>diagnosis <strong>of</strong> nasopharyngeal carcinoma. <strong>Head</strong> <strong>Neck</strong> 17:487–493120. Maher A, Bassiouny A, Bucci TJ, Moawad MK, Hendawy DS(1982) Tonsillomycosis: a myco-histopathological study. JLaryngol Otol 96:229–240121. McEwan J, Basha S, Rogers S, Harkness P (2001) An unusualpresentation <strong>of</strong> cat-scratch disease. J Laryngol Otol 115:826–828122. Mentzel T, Bainbridge TC, Katenkamp D (1997) Solitary fibroustumour: clinicopathological, immunohistochemical,<strong>and</strong> ultrastructural analysis <strong>of</strong> 12 cases arising in s<strong>of</strong>t tissues,nasal cavity <strong>and</strong> nasopharynx, urinary bladder <strong>and</strong> prostate.Virchows Arch 430:445–453123. Mey AGL van der, Krieken JHJM van, Dulken H van, StetersAP van, Vielvoye J, Hulsh<strong>of</strong> JH (1989) Large pituitary adenomaswith extension into <strong>the</strong> nasopharynx. Report <strong>of</strong> threecases with a review <strong>of</strong> <strong>the</strong> literature. Ann Otol Rhinol Laryngol98:618–624124. Michaels L, Hyams VJ (1979) Amyloid in localised deposits<strong>and</strong> plasmacytomas <strong>of</strong> <strong>the</strong> respiratory tract. J Pathol 128:29–38125. Michal M, Sokol L, Mukensnabl P (1996) Salivary gl<strong>and</strong> anlagetumor. A case with widespread necrosis <strong>and</strong> large cystformation. <strong>Pathology</strong> 28:128–130126. Miglets AW, Barton CL (1970) Sarcoid <strong>of</strong> <strong>the</strong> tonsil. ArchOtolaryngol 92:516–517127. Miglets AW, Viall JH, Kataria YP (1977) Sarcoidosis <strong>of</strong> <strong>the</strong>head <strong>and</strong> neck. Laryngoscope 87:2038–2048128. Miller R, Sneed W (1985) Tornwaldt’s bursa. Clin Otolaryngol10:21–25129. Mitchell TE, Girling AC (1996) Hairy polyp <strong>of</strong> <strong>the</strong> tonsil. JLaryngol Otol 110:101–103130. Miyahara H, Matsunaga T (1994) Tornwaldt’s disease. ActaOtolaryngol Suppl 517:36–39131. Moar E, Tovi F, Sacks M (1983) Carcinoma <strong>of</strong> <strong>the</strong> pancreaspresenting with bilateral tonsillar metastases. Ann Otol RhinolLaryngol 92:192–195132. Möller P, Wirbel R, H<strong>of</strong>mann W (1984) Lymphoepi<strong>the</strong>lialcarcinoma (Schmincke type) as a derivate <strong>of</strong> <strong>the</strong> tonsillarcrypt epi<strong>the</strong>lium. Virchows Arch (Pathol Anat) 405:85–93133. Monem S, O’Connor P, O’Leary T (1999) Peritonsillar abscess<strong>and</strong> infectious mononucleosis: an association or a differentpresentation <strong>of</strong> <strong>the</strong> same condition. Ir Med J 92:278–280134. Moore KL, Persaud TVN (1993) The developing human: clinicallyoriented embryology, 5th edn. Saunders, Philadelphia135. Mori N, Yatabe Y, Oka K, Kinoshita T, Kobayashi T, Ono T,Asai J (1996) Expression <strong>of</strong> perforin in nasal lymphoma. Additionalevidence <strong>of</strong> its natural killer cell derivation. Am JPathol 149:699–705136. Morin GV, Shank C, Burgess PA, Heffner DK (1991) Oncocyticmetaplasia <strong>of</strong> <strong>the</strong> pharynx. Otolaryngol <strong>Head</strong> <strong>Neck</strong>Surg 105:86137. Morrison D, Rose EL, Smith AP, Lesser TH (1992) Dyskeratosiscongenita <strong>and</strong> nasopharyngeal atresia. J Laryngol Otol106:996–997138. Müller E, Beleites E (2000) The basaloid squamous cell carcinoma<strong>of</strong> <strong>the</strong> nasopharynx. Rhinology 38:208–211139. Nageris B, Feinmesser M, Brama I, Feinmesser R (1991) Liposarcoma<strong>of</strong> <strong>the</strong> nasopharynx: a case report. Ear Nose ThroatJ 70:520–522140. Nave H, Gebert A, Pabst R (2001) Morphology <strong>and</strong> immunology<strong>of</strong> <strong>the</strong> human palatine tonsil. Anat Embryol 204:367–373141. Neel 3rd HB (1985) Nasopharyngeal carcinoma. Clinicalpresentation, diagnosis, treatment, <strong>and</strong> prognosis. OtolaryngolClin North Am 18:479–490142. Ng CS, Lo ST, Chan JK, Chan WC (1997) CD56+ putativenatural killer cell lymphomas: production <strong>of</strong> cytolytic effectors<strong>and</strong> related proteins mediating tumor cell apoptosis?Hum Pathol 28:1276–1282143. Nicolai P, Luzzago F, Maroldi R, Falchetti M, Antonelli A(1989) Nasopharyngeal cysts. Report <strong>of</strong> seven cases with review<strong>of</strong> <strong>the</strong> literature. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg115:860–864144. Nojeg MM, Jalaludin MA, Jayalakshmi P (1998) Papillaryadenocarcinoma <strong>of</strong> <strong>the</strong> nasopharynx – case report <strong>and</strong> review<strong>of</strong> <strong>the</strong> literature. Med J Malaysia 53:104–106145. Norval E (1998) A mucosa-associated lymphoid tissue(MALT) type lymphoma (marginal zone B-cell lymphoma)from <strong>the</strong> palatine tonsil. <strong>Pathology</strong> 30:209–211146. Norval E, Thompson I (2001) Non-Hodgkin’s lymphomas <strong>of</strong>Waldeyer’s ring: a clinicopathological <strong>and</strong> immunologicalstudy <strong>of</strong> 64 cases in <strong>the</strong> western Cape. SADJ 56:545–548147. Nouwen J, Smets F, Rombaux P, Hamoir M, Sokal EM (2002)Acute tonsillitis as <strong>the</strong> first manifestation <strong>of</strong> post-transplantlymphoproliferative disorder. Ann Otol Rhinol Laryngol111:165–168148. Ohshima K, Suzumiya J, Shimazaki K, Kato A, Tanaka T,K<strong>and</strong>a M, Kikuchi M (1997) Nasal T/NK cell lymphomascommonly express perforin <strong>and</strong> Fas lig<strong>and</strong>: important mediators<strong>of</strong> tissue damage. Histopathology 31:444–450149. Ol<strong>of</strong>sson K, Hellstrom S, Hammarstrom M (1998) The surfaceepi<strong>the</strong>lium <strong>of</strong> recurrent infected palatine tonsils is richin gammadelta T cells. Clin Exp Immunol 111:36–47150. Ott G, Kalla J, Ott MM, Müller-Hermelink HK (1997) TheEpstein-Barr virus in malignant non-Hodgkin’s lymphoma<strong>of</strong> <strong>the</strong> upper aerodigestive tract. Mol Pathol 6:134–139151. Oudejans JJ, Harijadi H, Kummer JA, Tan IB, Bloemena E,Middele JM, Bladergroen B, Dukers DF, Vos W, Meijer CJ(2002) High numbers <strong>of</strong> granzyme B/CD8-positive tumourinfiltratinglymphocytes in nasopharyngeal carcinoma biopsiespredict rapid fatal outcome in patients treated with curativeintent. J Pathol 198:468–475152. Papadas T, Batistatou A, Ravazoula P, Zolota V, Goumas P(2001) S-100 protein-positive dendritic cells <strong>and</strong> CD-34 positivedendritic interstitial cells in palatine tonsils. Arch Otorhinolaryngol258:243–245153. Paris J, Guelfucci B, Moulin G, Zanaret M, Triglia JM (2001)Diagnosis <strong>and</strong> treatment <strong>of</strong> juvenile nasopharyngeal angi<strong>of</strong>ibroma.Eur Arch Otorhinolaryngol 258:120–124


Nasopharynx <strong>and</strong> Waldeyer’s Ring Chapter 6 195154. Parks SS, Karmody CS (1992) The first branchial cleft carcinoma.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 118:969–971155. Paslin D (1980) Accessory tonsils. Arch Dermatol 116:720–721156. Patel A, Pambuccian S, Maisel R (2002) Nasopharyngeal amyloidosis.Am J Otolaryngol 23:308–311157. Pathmanathan R, Prasad U, Ch<strong>and</strong>rika G, Sadler R, FlynnK, Raab-Traub N (1995) Undifferentiated, nonkeratinizing,<strong>and</strong> squamous cell carcinoma <strong>of</strong> <strong>the</strong> nasopharynx. Variants<strong>of</strong> Epstein-Barr virus-infected neoplasia. Am J Pathol146:1355–1367158. Pathmanathan R, Prasad U, Sadler R, Flynn K, Raab-TraubN (1995) Clonal proliferations <strong>of</strong> cells infected with Epstein-Barr virus in preinvasive lesions related to nasopharyngealcarcinoma. N Engl J Med 333:693–698159. Paulino AF, Singh B, Shah JP, Huvos AG (2000) Basaloidsquamous cell carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Laryngoscope110:1479–1482160. Paz IB, Cook N, Odom-Maryon T, Xie Y, Wilczynski SP(1997) Human papillomavirus (HPV) in head <strong>and</strong> heck cancer.An association <strong>of</strong> HPV 16 with squamous cell carcinoma<strong>of</strong> Waldeyer’s tonsillar ring. Cancer 79:595–604161. Percodani J, Braun F, Arrue P, Yardeni E, Murris-Espin M,Serrano, Pessey JJ (1999) Nasopharyngeal tuberculosis. JLaryngol Otol 113:928–931162. Perzin KH, Pushparaj N (1984) Nonepi<strong>the</strong>lial tumors <strong>of</strong> <strong>the</strong>nasal cavity, paranasal sinuses, <strong>and</strong> nasopharynx. A clinicopathologicstudy. XIII: meningiomas. Cancer 54:1860–1869163. Posey LA, Kerschner JE, Conley SF (1999) Posttransplantationlymphoproliferative disease in children: otolaryngologicmanifestations <strong>and</strong> management. South Med J 92:1079–1082164. Reddy S, Raslan W, Gooneratne S, Kathuria S, Marks J (1995)Prognostic significance <strong>of</strong> keratinization in nasopharyngealcarcinoma. Am J Otolaryngol 16:103–108165. Regauer S, Beham A, Mannweiler S (2000) CK7 expression incarcinomas <strong>of</strong> <strong>the</strong> Waldeyer’s ring area. Hum Pathol 31:1096–1101166. Regauer S, Mannweiler S, Anderhuber W, Gottschuli A,Berghold A, Schachenreiter J, Jakse R, Beham A (1999) Cysticlymph node metastases <strong>of</strong> squamous cell carcinoma <strong>of</strong>Waldeyer’s ring origin. Br J Cancer 1437–1442167. Rowe LD (1980) Neonatal airway obstruction secondaryto nasopharyngeal teratoma. Orolaryngol <strong>Head</strong> <strong>Neck</strong> Surg88:221–226168. Ruff T, Diaz JA (1986) Heterotopic brain in <strong>the</strong> nasopharynx.Orolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 94:254–256169. Sabo R, Sela M, Sabo G, Herskovitz P, Feinmesser R (2001)Metastatic hypernephroma to <strong>the</strong> head <strong>and</strong> neck: unusualcase reports <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. J Otolaryngol30:140–144170. Schick B, Brunner C, Praetorius M, Plinkert PK, UrbschatS (2002) First evidence <strong>of</strong> genetic imbalances in angi<strong>of</strong>ibromas.Laryngoscope 112:397–401171. Schick B, Plinkert PK, Prescher A (2002) Aetiology <strong>of</strong> angi<strong>of</strong>ibromas:reflection on <strong>the</strong>ir specific vascular component.Laryngorhinootologie 81:280–284172. Scott A, Stansbie JM (1997) Actinomycosis presenting asa nasopharyngeal tumour: a case report. J Laryngol Otol111:163–169173. Sellars SL (1971) Metastatic tumours <strong>of</strong> <strong>the</strong> tonsil. J LaryngolOtol 85:289–292174. Shahab I, Osborne B, Butler J (1994) Nasopharyngeal lymphoidtissue masses in patients with human immunodeficiencyvirus-1. Cancer 74:3083–3088175. Sham J, Poon YF, Wei WI, Choy D (1990) Nasopharyngealcarcinoma in young patients. Cancer 65:2606–2610176. Shanmugaratnam K, Sobin LH (1991) Histological typing <strong>of</strong>tumours <strong>of</strong> <strong>the</strong> upper respiratory tract <strong>and</strong> ear. WHO InternationalHistological Classification <strong>of</strong> Tumours. Springer,Berlin Heidelberg New York177. Shidara K, Uruma T, Yasuoka Y, Kamei T (1993) Two cases<strong>of</strong> nasopharyngeal branchial cyst. J Laryngol Otol 107:453–455178. Shreedhar R, Tooley AH (1984) Carcinoma arising in a branchialcyst. Br J Surg 75:115179. Siebert JD, Stuckey JH, Kurtin PJ, Banks PM (1995) Extranodallymphocyte predominance Hodgkin’s disease. Clinical<strong>and</strong> pathologic features. Am J Clin Pathol 103:485–491180. Smi<strong>the</strong>ringale A (2000) Lymphomas presenting in Waldeyer’sring. Otolaryngology 29:183–185181. S<strong>of</strong>ferman RA, Heisse JWH Jr (1985) Adenoid cystic carcinoma<strong>of</strong> <strong>the</strong> nasopharynx after previous adenoid irradiation.Laryngoscope 95:458–461182. Strickler J, Fedeli F, Horowitz CA, Copenhaver CM, FrizzeraG (1993) Infectious mononucleosis in lymphoid tissue. Histopathology,in-situ hybridization, <strong>and</strong> differential diagnosis.Arch Pathol Lab Med 117:269–278183. Sulica RL, Wenig BM, Debo RF, Sessions RB (1999) Schneiderianpapillomas <strong>of</strong> <strong>the</strong> pharynx. Ann Otol Rhinol Laryngol108:392–397184. Taddasse-Heath L, Feldman J, Fahle GA, Fisher SH, SorbaraL, Raffeld M, Jaffe E (2003) Florid CD4+, CD56+ T-cell infiltrateassociated with Herpes simplex infection simulatingnasal NK-/T-cell lymphoma. Mod Pathol 16:166–172185. Tamagawa Y, Kitamura K, Miyata M (1995) Branchial cyst<strong>of</strong> <strong>the</strong> nasopharynx: resection via <strong>the</strong> endonasal approach. JLaryngol Otol 109:139–141186. Tao Q, Srivastava G, Chan AC, Chung LP, Loke SL, Ho FC(1995) Evidence for lytic infection by Epstein-Barr virus inmucosal lymphocytes instead <strong>of</strong> nasopharyngeal epi<strong>the</strong>lialcells in normal individuals. J Med Virol 45:71–77187. Tharrington C, Bossen E (1992) Nasopharyngeal teratomas.Arch Pathol Lab Med 116:165–167188. Thompson L, Gyure K (2000) Extracranial sinonasal tractmeningiomas. a clinicopathologic study <strong>of</strong> 30 cases with areview <strong>of</strong> <strong>the</strong> literature. Am J Surg Pathol 24:640–650189. Thompson LDR, Heffner DK (1998) The clinical importance<strong>of</strong> cystic squamous cell carcinomas in <strong>the</strong> neck. Cancer82:944–956190. Tominaga S, Fukushima K, Nishizaki K, Watanabe S, MasudaY, Ogura H (1996) Presence <strong>of</strong> human papillomavirustype 6 in tonsillar condyloma acuminatum <strong>and</strong> clinicallynormal tonsillar mucosa. Jpn J Clin Oncol 26:393–397191. Tsai ST, Jin YT, Su IJ (1996) Expression <strong>of</strong> EBER1 in primary<strong>and</strong> metastatic nasopharyngeal carcinoma tissues using insitu hybridization. A correlation with WHO histologic subtypes.Cancer 77:231–236192. Voelker JL, Campell RL, Muller L (1991) Clinical, radiographic<strong>and</strong> pathological features <strong>of</strong> symptomatic Rathke’scleft cysts. J Neurosurg 74:535–644193. Waldron J, Hasselt CAV, Skinner DW, Arnold M (1992) Tuberculosis<strong>of</strong> <strong>the</strong> nasopharynx: clinicopathological features.Clin Otolaryngol 17:57–59194. Wan SK, Chan JK, Lau WH, Yip TT (1995) Basaloid-squamouscarcinoma <strong>of</strong> <strong>the</strong> nasopharynx. An Epstein-Barr virusassociatedneoplasm compared with morphologically identicaltumors occurring in o<strong>the</strong>r sites. Cancer 76:1689–1693195. Wat PJ, Strickler JG, Jeffrey L, Nordstrom MR (1994) Herpessimplex infection causing acute necrotizing tonsillitis. MayoClin Proc 69:269–271196. Weiss LM, Movahed LA, Butler AE, Swanson SA, FriersonHFJ, Cooper PH, Colby TV, Mills SE (1989) Analysis <strong>of</strong> lymphoepi<strong>the</strong>lioma<strong>and</strong> lymphoepi<strong>the</strong>lioma-like carcinomas forEpstein-Barr vital genomes by in situ hybridization. Am JSurg Pathol 13:625–631197. Wenig B, Thompson L, Frankel S (1996) Lymphoid changes<strong>of</strong> <strong>the</strong> nasopharyngeal <strong>and</strong> palatine tonsils that are indicative<strong>of</strong> human immunodeficiency virus infection. Am J SurgPathol 20:572–587198. Wenig BM, Harpaz N, DelBridge C (1989) Polymorphouslow-grade adenocarcinoma <strong>of</strong> seromucous gl<strong>and</strong>s <strong>of</strong> <strong>the</strong> nasopharynx.A report <strong>of</strong> a case <strong>and</strong> a discussion <strong>of</strong> <strong>the</strong> morphologic<strong>and</strong> immunohistochemical features. Am J ClinPathol 92:104–109199. Wenig BM, Heffner DK (1995) Respiratory epi<strong>the</strong>lial adenomatoidhamartomas <strong>of</strong> <strong>the</strong> sinonasal tract <strong>and</strong> nasopharynx:a clinicopathologic study <strong>of</strong> 31 cases. Ann Otol Rhinol Laryngol104:639–645


196 S. Regauer200. Win<strong>the</strong>r B, Innes D (1994) The human adenoid. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 120:144–149201. Witkin GB, Rosai J (1991) Solitary fibrous tumor <strong>of</strong> <strong>the</strong> upperrespiratory tract. A report <strong>of</strong> six cases. Am J Surg Pathol15:842–848202. Woodruff C (1980) Nonneoplastic <strong>and</strong> inflammatory disorders<strong>of</strong> Waldeyer’s ring. Ear Nose Throat J 59:457–465203. Yadav SP, Singh I, Ch<strong>and</strong>a R, Sachdeva OP (2002) Nasopharyngealangi<strong>of</strong>ibroma. Otolaryngol 31:346–350204. Yamanaka N, Kataura A (1984) Viral infections associatedwith recurrent tonsillitis. Acta Otolaryngol Suppl 416:30–37205. Ylikoski J, Karjalainen J (1989) Acute tonsillitis in youngmen: etiological agents <strong>and</strong> <strong>the</strong>ir differentiation, Sc<strong>and</strong> J Infect21:169–174206. Zarbo R, McClatchey K (1983) Nasopharyngeal hamartoma.Laryngoscope 93:494–497207. Ziegler JL, Beckstead JA, Volberding PA, Abrams DI, LevineAM, Lukes RJ, Gill PS, Burkes RL, Meyer PR, Metroka CE(1984) Non-Hodgkin’s lymphoma in 90 homosexual men.Relation to generalized lymphadenopathy <strong>and</strong> <strong>the</strong> acquiredimmunodeficiency syndrome. N Engl J Med 311:565–570208. Zong Y, Zhang C, Zhang F, Ruan JB, Chen MY, Feng KT, YuZF (1993) Infiltrating lymphocytes <strong>and</strong> accessory cells in nasopharyngealcarcinoma. Jpn J Cancer Res 84:900–9056


Chapter 7Larynx<strong>and</strong> HypopharynxN. Gale · A. Cardesa · N. Zidar7Contents7.1 Summary <strong>of</strong> Anatomy,Histology <strong>and</strong> Embryology . . . . . . . . . . . . . . . . 1987.2 Laryngocele, Cysts, Heterotopia . . . . . . . . . . . . . 1997.2.1 General Considerations . . . . . . . . . . . . . . . . . 1997.2.2 Laryngocele . . . . . . . . . . . . . . . . . . . . . . . . 1997.2.3 Sacccular Cyst . . . . . . . . . . . . . . . . . . . . . . . 1997.2.4 Ductal Cyst . . . . . . . . . . . . . . . . . . . . . . . . . 1997.2.5 Oncocytic Cyst . . . . . . . . . . . . . . . . . . . . . . . 2007.2.6 Zenker’s Hypopharyngeal Diverticle . . . . . . . . . . 2017.2.7 Aberrant Thyroid Tissue . . . . . . . . . . . . . . . . . 2017.2.8 Tracheopathia Osteochondroplastica . . . . . . . . . 2027.3 Inflammatory Lesions . . . . . . . . . . . . . . . . . . 2027.3.1 Acute Infections . . . . . . . . . . . . . . . . . . . . . . 2027.3.1.1 Epiglottitis . . . . . . . . . . . . . . . . . . . . . . . . . 2027.3.1.2 Laryngotracheobronchitis . . . . . . . . . . . . . . . . 2027.3.1.3 Diph<strong>the</strong>ria . . . . . . . . . . . . . . . . . . . . . . . . . 2027.3.2 Chronic Infections . . . . . . . . . . . . . . . . . . . . 2027.3.2.1 Tuberculosis . . . . . . . . . . . . . . . . . . . . . . . . 2027.3.2.2 Fungal Infections . . . . . . . . . . . . . . . . . . . . . 2037.3.2.3 O<strong>the</strong>r Rare Infections . . . . . . . . . . . . . . . . . . . 2037.3.3 Non-Infectious Inflammatory Lesions . . . . . . . . . 2037.3.3.1 Wegener’s Granulomatosis . . . . . . . . . . . . . . . . 2037.3.3.2 Sarcoidosis . . . . . . . . . . . . . . . . . . . . . . . . . 2047.3.3.3 Rheumatoid Arthritis . . . . . . . . . . . . . . . . . . . 2047.3.3.4 Relapsing Polychondritis . . . . . . . . . . . . . . . . . 2057.3.3.5 Gout . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2067.3.3.6 Teflon Granuloma . . . . . . . . . . . . . . . . . . . . . 2067.3.3.7 Idiopathic Subglottic Laryngeal Stenosis . . . . . . . 2067.3.3.8 Angioneurotic Oedema . . . . . . . . . . . . . . . . . . 2077.4 Degenerative Lesions . . . . . . . . . . . . . . . . . . . 2077.4.1 Oculopharyngeal Muscular Dystrophy . . . . . . . . 2077.5 Pseudotumours . . . . . . . . . . . . . . . . . . . . . . 2077.5.1 Exudative Lesions <strong>of</strong> Reinke’s Space . . . . . . . . . . 2077.5.1.1 Reinke’s Oedema . . . . . . . . . . . . . . . . . . . . . 2087.5.1.2 Vocal Cord Polyp <strong>and</strong> Nodule . . . . . . . . . . . . . . 2087.5.2 Contact Ulcer <strong>and</strong> Granuloma,Intubation Granuloma . . . . . . . . . . . . . . . . . . 2107.5.3 Necrotising Sialometaplasia . . . . . . . . . . . . . . . 2117.5.4 Metaplastic Elastic Cartilaginous Nodules . . . . . . 2117.5.5 Amyloidosis . . . . . . . . . . . . . . . . . . . . . . . . 2117.5.6 Sinus Histiocytosis with Massive Lymphadenopathy<strong>and</strong> O<strong>the</strong>r Rare Pseudotumours . . . . . . . . . . . . . 2127.5.7 Inflammatory My<strong>of</strong>ibroblastic Tumour . . . . . . . . 2137.6 Benign Neoplasms . . . . . . . . . . . . . . . . . . . . . 2147.6.1 Squamous Cell Papilloma . . . . . . . . . . . . . . . . 2147.6.2 Salivary Gl<strong>and</strong>-Type Tumours . . . . . . . . . . . . . . 2147.6.2.1 Pleomorphic Adenoma . . . . . . . . . . . . . . . . . . 2147.6.2.2 Oncocytoma . . . . . . . . . . . . . . . . . . . . . . . . 2147.6.3 Haemangioma (Neonatal <strong>and</strong> Adult Types) . . . . . . 2147.6.4 Paraganglioma . . . . . . . . . . . . . . . . . . . . . . . 2157.6.5 Granular Cell Tumour . . . . . . . . . . . . . . . . . . 2167.6.6 Chondroma . . . . . . . . . . . . . . . . . . . . . . . . . 2177.7 Malignant Neoplasms . . . . . . . . . . . . . . . . . . . 2177.7.1 Potentially Malignant (Precancerous) Lesions . . . . 2177.7.2 Invasive Squamous Cell Carcinoma . . . . . . . . . . 2187.7.2.1 Epidemiology . . . . . . . . . . . . . . . . . . . . . . . 2187.7.2.2 Aetiology . . . . . . . . . . . . . . . . . . . . . . . . . . 2187.7.2.3 Anatomic Sites . . . . . . . . . . . . . . . . . . . . . . . 2187.7.2.4 Histological Variants . . . . . . . . . . . . . . . . . . . 2197.7.2.5 TNM Grading . . . . . . . . . . . . . . . . . . . . . . . 2207.7.3 Neuroendocrine Carcinoma . . . . . . . . . . . . . . . 2207.7.3.1 Well-Differentiated NeuroendocrineCarcinoma (Carcinoid) . . . . . . . . . . . . . . . . . . 2207.7.3.2 Moderately Differentiated NeuroendocrineCarcinoma (Atypical Carcinoid) . . . . . . . . . . . . 2207.7.3.3 Poorly Differentiated NeuroendocrineCarcinoma (Small Cell Carcinoma) . . . . . . . . . . 2217.7.4 Adenocarcinoma . . . . . . . . . . . . . . . . . . . . . 2227.7.4.1 Adenoid Cystic Carcinoma . . . . . . . . . . . . . . . 2227.7.4.2 Mucoepidermoid Carcinoma . . . . . . . . . . . . . . 2227.7.5 Sarcomas . . . . . . . . . . . . . . . . . . . . . . . . . . 2237.7.5.1 Chondrosarcoma . . . . . . . . . . . . . . . . . . . . . 2237.7.5.2 O<strong>the</strong>r Sarcomas . . . . . . . . . . . . . . . . . . . . . . 2247.7.6 O<strong>the</strong>r Malignant Neoplasms . . . . . . . . . . . . . . . 2247.7.6.1 Malignant Lymphoma . . . . . . . . . . . . . . . . . . 2247.7.6.2 Extraosseus (Extramedullary) Plasmacytoma . . . . . 2247.7.6.3 Primary Mucosal Melanoma . . . . . . . . . . . . . . . 2257.7.6.4 Metastases to <strong>the</strong> Larynx . . . . . . . . . . . . . . . . . 225References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226


198 N. Gale · A. Cardesa · N. Zidar77.1 Summary <strong>of</strong> Anatomy,Histology <strong>and</strong> EmbryologyThe larynx <strong>and</strong> hypopharynx are constituent parts <strong>of</strong><strong>the</strong> upper aerodigestive tract, intimately linked toge<strong>the</strong>rwith <strong>the</strong>ir connective tissue elements <strong>and</strong> differentepi<strong>the</strong>lia. The anatomy <strong>and</strong> histology <strong>of</strong> both organsare very complex <strong>and</strong> details are available in variousst<strong>and</strong>ard textbooks <strong>and</strong> specialised papers [103, 158,238, 321, 322]. Only essential data will be given here.The larynx is a hollow tube that communicatescranially with <strong>the</strong> hypopharynx. Its upper limits are<strong>the</strong> free edge <strong>of</strong> <strong>the</strong> epiglottis <strong>and</strong> <strong>the</strong> two aryepiglotticfolds. The lower laryngeal part continues caudallywith <strong>the</strong> trachea, <strong>and</strong> its inferior limit is <strong>the</strong> loweredge <strong>of</strong> <strong>the</strong> cricoid cartilage. The anterior border consists<strong>of</strong> <strong>the</strong> lingual surface <strong>of</strong> <strong>the</strong> epiglottis, thyrohyoidmembrane, thyroid cartilage <strong>and</strong> <strong>the</strong> anterior arch <strong>of</strong><strong>the</strong> cricoid cartilage. Posteriorly, <strong>the</strong> cricoid cartilage<strong>and</strong> area <strong>of</strong> <strong>the</strong> arytenoids limit <strong>the</strong> larynx.The larynx is divided into supraglottic, glottic <strong>and</strong>subglottic regions, which have particular significancefor <strong>the</strong> biological behaviour <strong>and</strong> staging <strong>of</strong> tumours.The supraglottic region extends from <strong>the</strong> tip <strong>of</strong> <strong>the</strong> epiglottisdown to <strong>the</strong> superior edge <strong>of</strong> <strong>the</strong> true vocal cord<strong>and</strong> includes <strong>the</strong> epiglottis, aryepiglottic folds, arytenoids,<strong>the</strong> false vocal cords <strong>and</strong> ventricles. The glottisincludes <strong>the</strong> vocal cords with anterior <strong>and</strong> posteriorcommissures. The subglottis extends below <strong>the</strong>true vocal cords to <strong>the</strong> lower border <strong>of</strong> <strong>the</strong> cricoid cartilage.Our own experience suggests that marked variationsin <strong>the</strong> distribution <strong>of</strong> different types <strong>of</strong> laryngeal epi<strong>the</strong>liarelated to age seem to be <strong>the</strong> rule [177]. The lingual<strong>and</strong>, variably, laryngeal side <strong>of</strong> <strong>the</strong> epiglottis <strong>and</strong><strong>the</strong> true vocal cords are covered by <strong>the</strong> non-keratinisingstratified squamous cell epi<strong>the</strong>lium, <strong>the</strong> rest <strong>of</strong> <strong>the</strong>larynx is lined with <strong>the</strong> respiratory epi<strong>the</strong>lium. The seromucinousgl<strong>and</strong>s are abundant in all compartments<strong>of</strong> <strong>the</strong> larynx except in <strong>the</strong> vocal cords, where <strong>the</strong>y areessentially missing on <strong>the</strong>ir free edges <strong>and</strong> are sparse in<strong>the</strong> rest <strong>of</strong> <strong>the</strong> cords.It is important to draw attention to some particularitiesin <strong>the</strong> laryngeal structure that considerably influence<strong>the</strong> spread <strong>of</strong> malignant tumours. The elasticcartilage <strong>of</strong> <strong>the</strong> epiglottis with numerous fenestrationsfor vessels, nerves <strong>and</strong> gl<strong>and</strong>s provides a “locus minorisresistentiae” for <strong>the</strong> progress <strong>of</strong> malignant growthfrom <strong>the</strong> laryngeal to <strong>the</strong> lingual side or vice versa. In<strong>the</strong> anterior commissure, where <strong>the</strong> true vocal cordsmeet in <strong>the</strong> anterior midline, a b<strong>and</strong> <strong>of</strong> fibrous tissue(protrusions <strong>of</strong> <strong>the</strong> two vocal ligaments) with lymphatic<strong>and</strong> blood vessels is attached to <strong>the</strong> thyroid cartilage.There is no perichondrium at this point, whichcertainly facilitates <strong>the</strong> ingrowth <strong>of</strong> malignant tu-mours in <strong>the</strong> thyroid cartilage. The network <strong>of</strong> capillariesis poorly developed in Reinke’s space <strong>of</strong> <strong>the</strong> vocalcords <strong>and</strong> lymphatics are lacking. These specificitiescontribute to <strong>the</strong> development <strong>of</strong> various exudativelesions <strong>of</strong> <strong>the</strong> vocal cords <strong>and</strong> delayed metastases<strong>of</strong> glottic cancers.Embryologically, <strong>the</strong> supraglottic part <strong>of</strong> <strong>the</strong> larynxarises from <strong>the</strong> third <strong>and</strong> <strong>the</strong> fourth branchial arches,while <strong>the</strong> glottic <strong>and</strong> subglottic portions are derivedfrom <strong>the</strong> sixth arch. The first appearance <strong>of</strong> <strong>the</strong> respiratorytract occurs at approximately 21 days duringembryogenesis as an evagination or a vertical groove<strong>of</strong> <strong>the</strong> cephalic portion <strong>of</strong> <strong>the</strong> foregut. This evaginationis <strong>the</strong> precursor <strong>of</strong> <strong>the</strong> epiglottis, <strong>the</strong> earliest portion <strong>of</strong><strong>the</strong> larynx. Its outlines appear at <strong>the</strong> 6-mm foetal stageby 30 days. The respiratory groove begins to close <strong>and</strong>with <strong>the</strong> formation <strong>of</strong> <strong>the</strong> arytenoids, <strong>the</strong> closure becomescomplete [103]. The covering epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong>groove appears in <strong>the</strong> 3–5-mm embryo as three lines <strong>of</strong>polyhedral embryonic cells <strong>of</strong> endodermal origin. In a30-mm foetus, by 60–70 days, <strong>the</strong> thickness <strong>of</strong> <strong>the</strong> embryonicstratified squamous epi<strong>the</strong>lium increases, <strong>and</strong><strong>the</strong> vocal cords begin to differentiate. The ciliated epi<strong>the</strong>liumoccurs in a 40-mm foetus on <strong>the</strong> epiglottis <strong>and</strong>laryngeal vestibule. A sharp distinction between <strong>the</strong>two epi<strong>the</strong>lia appears after <strong>the</strong> foetus reaches a length<strong>of</strong> 95 mm. The larynx <strong>of</strong> a newborn is covered in <strong>the</strong>ciliated epi<strong>the</strong>lium, except on <strong>the</strong> true vocal cords. Inaddition to this location, <strong>the</strong> stratified squamous epi<strong>the</strong>liumis also present on <strong>the</strong> interarytenoid area <strong>and</strong>on <strong>the</strong> tip <strong>of</strong> <strong>the</strong> epiglottis [158].The hypopharynx is <strong>the</strong> caudal part <strong>of</strong> <strong>the</strong> pharynxwith <strong>the</strong> wide part superiorly, extending from <strong>the</strong> tip<strong>of</strong> <strong>the</strong> epiglottis to <strong>the</strong> inferior level <strong>of</strong> <strong>the</strong> cricoid cartilage,where it becomes narrow <strong>and</strong> continuously proceedsto <strong>the</strong> oesophagus. The hypopharynx is dividedinto three compartments: left <strong>and</strong> right pyriform sinuses,postcricoidal region, <strong>and</strong> posterior pharyngealwalls. The pyriform sinuses are medially limited by<strong>the</strong> aryepiglottic folds <strong>and</strong> laterally by <strong>the</strong> thyroid cartilage.The postcricoid area is <strong>the</strong> posterior side <strong>of</strong> <strong>the</strong>cricoid cartilage. The posterior wall is situated in front<strong>of</strong> <strong>the</strong> cervical spine. The entire hypopharynx is coveredin <strong>the</strong> stratified squamous cell epi<strong>the</strong>lium.Embryologically, <strong>the</strong> pharyngeal gut or pharynx extendsfrom <strong>the</strong> buccopharyngeal membrane to <strong>the</strong> tracheobronchialdiverticulum. The hypopharynx is almostentirely <strong>of</strong> endodermal origin. In <strong>the</strong> 8th throughto <strong>the</strong> 10th gestation week <strong>the</strong> pharynx, as well as <strong>the</strong>hypopharynx, is ra<strong>the</strong>r small <strong>and</strong> after <strong>the</strong> 10th week<strong>of</strong> gestation, remarkable growth occurs in this region[204, 252].


Larynx <strong>and</strong> Hypopharynx Chapter 7 1997.2 Laryngocele,Cysts, Heterotopia7.2.1 General ConsiderationsA laryngocele is a rare congenital or acquired laryngeallesion that appears within <strong>and</strong> around <strong>the</strong> laryngeal saccule.Laryngeal cysts account for approximately 5% <strong>of</strong>benign laryngeal lesions [177, 265]. DeSanto presenteda classification <strong>of</strong> laryngeal cysts in which <strong>the</strong> lesionswere divided into ductal, saccular <strong>and</strong> thyroid cartilageforaminal cysts [80]. This classification, which ismore clinically adjusted, is based on <strong>the</strong> intramucosaldepth <strong>of</strong> <strong>the</strong> cyst <strong>and</strong> its location. Newman <strong>and</strong> coworkersfound it difficult to apply <strong>and</strong> proposed a newone, dividing <strong>the</strong> lesions into tonsillar, epi<strong>the</strong>lial (saccular<strong>and</strong> ductal) <strong>and</strong> oncocytic cysts. According to <strong>the</strong>original article, more than half <strong>of</strong> all laryngeal cystswere epi<strong>the</strong>lial, one quarter tonsillar <strong>and</strong> less than 15%oncocytic [265].7.2.2 LaryngoceleLaryngocele is defined as an excessive elongation <strong>and</strong>dilatation <strong>of</strong> <strong>the</strong> air-filled laryngeal saccule (ventricularappendix), which communicates directly with <strong>the</strong> laryngeallumen. According to its site, <strong>the</strong>re are three types <strong>of</strong>lesions: internal, external, <strong>and</strong> mixed. An internal laryngoceleextends in a superior-posterior direction, towards<strong>the</strong> area <strong>of</strong> <strong>the</strong> false vocal cord <strong>and</strong> aryepiglottic fold. Anexternal one exp<strong>and</strong>s cranially <strong>and</strong> laterally to <strong>the</strong> neckthrough <strong>the</strong> weak zone <strong>of</strong> <strong>the</strong> thyrohyoid membrane.It presents as a lateral neck mass that varies in size dependingon variations <strong>of</strong> <strong>the</strong> intralaryngeal pressure. Amixed or combined form has both internal <strong>and</strong> externalcomponents with a swelling <strong>of</strong> <strong>the</strong> neck <strong>and</strong> endolaryngealbulging [14, 15, 53]. The combined laryngocele is<strong>the</strong> most common (44%), followed by internal (30%) <strong>and</strong>external (26%) forms [50].A laryngocele is quite a rare lesion, occurring ascongenital [60] or acquired, most frequently observedin infants <strong>and</strong> adults between 50 <strong>and</strong> 60 years. A malepredominance is evident with a ratio <strong>of</strong> 7:1 [227, 287].Most laryngoceles are unilateral. Aetiologically, <strong>the</strong>lesion occurs in persons with a congenital large saccule<strong>and</strong> weakness <strong>of</strong> <strong>the</strong> periventricular s<strong>of</strong>t tissue.In adults, various conditions involving a repeatedincrease in intralaryngeal pressure, such as inflictingglass-blowers, wind instrument musicians, singers,pr<strong>of</strong>essional speakers, <strong>and</strong> patients with a chroniccough, are reported [227]. Stenosis <strong>of</strong> <strong>the</strong> saccule neck,which functions as a valve system, may also lead to<strong>the</strong> occurrence <strong>of</strong> a laryngocele. The leading symptoms<strong>of</strong> an internal or compound lesion are hoarseness,cough, dyspnoea, dysphagia, <strong>and</strong> <strong>the</strong> sensation<strong>of</strong> a foreign body.The lesion may, however, be also asymptomatic in approximately12% <strong>of</strong> <strong>the</strong> cases [62]. The diagnosis is establishedby <strong>the</strong> history, <strong>and</strong> by physical <strong>and</strong> radiologicalexamination, especially computed tomography (CT).Histologically, a cystic extension <strong>of</strong> <strong>the</strong> saccule is evident<strong>and</strong> its wall tends to lose its folded surface. The laryngoceleis covered by <strong>the</strong> respiratory epi<strong>the</strong>lium; occasionallyan oncocytic or cuboidal metaplasia is present.Focally, chronic mononuclear inflammatory cellsare seen in <strong>the</strong> subepi<strong>the</strong>lial stroma. Laryngocele-relatedcomplications include infection (laryngopyocele),aspiration <strong>and</strong> subsequent pneumonia [287]. There isalso a relationship between laryngocele <strong>and</strong> laryngealsquamous cell carcinoma in 4.9 to 28.8% <strong>of</strong> cases [140].The endoscopic surgical treatment <strong>of</strong> laryngocele is <strong>the</strong>method <strong>of</strong> choice [354].7.2.3 Saccular CystA saccular cyst (SC) is a mucus-filled dilatation <strong>of</strong> <strong>the</strong>laryngeal saccule that has no communication with <strong>the</strong>laryngeal lumen [80, 161]. Most SCs are congenital inorigin; some may also appear as acquired lesions causedby various inflammatory processes, traumatic events,or tumours [1, 161, 257]. SCs, which may occur at anyage, are divided into anterior <strong>and</strong> lateral. The formerspread medially <strong>and</strong> posteriorly, <strong>and</strong> protrude into <strong>the</strong>laryngeal lumen between <strong>the</strong> true <strong>and</strong> false vocal cord.The latter are generally larger <strong>and</strong> extend towards <strong>the</strong>false vocal cord <strong>and</strong> aryepiglottic fold. They may rarelyspread through <strong>the</strong> thyrohyoid membrane [10, 80, 161,357]. SCs may be asymptomatic, but <strong>the</strong> most commonsymptoms are progressive cough, dysphagia, hoarseness,dyspnoea <strong>and</strong> foreign body sensation. Diagnosisis <strong>of</strong>ten made by laryngoscopy combined with CT scan[69].Histologically, SCs are lined with ciliated respiratoryepi<strong>the</strong>lium . An increased number <strong>of</strong> goblet cells may bepresent. Rarely, <strong>the</strong> cysts are partially or entirely linedby metaplastic squamous or oncocytic epi<strong>the</strong>lium. Subepi<strong>the</strong>lialstroma, i.e. <strong>the</strong> cyst wall, usually contains focallymphocytic infiltrates [177].Treatment is surgical, <strong>the</strong> decision on an endoscopicor an external approach depends on <strong>the</strong> type <strong>and</strong> size <strong>of</strong><strong>the</strong> cyst, as well as <strong>the</strong> individual patient’s condition.7.2.4 Ductal CystDuctal cysts (DCs) are <strong>the</strong> most common laryngeal cysts<strong>and</strong> comprise up to 62.5% <strong>of</strong> all laryngeal cystic lesions.The characteristic retention <strong>of</strong> mucus in <strong>the</strong> dilated collectingducts <strong>of</strong> <strong>the</strong> intramucosal seromucinous gl<strong>and</strong>s


200 N. Gale · A. Cardesa · N. Zidar7aFig. 7.1. Ductal cyst. a Large cyst arises from <strong>the</strong> right ventricle.b Ductal cyst is covered in ductal epi<strong>the</strong>lium <strong>and</strong> filled with mucinbcan be found anywhere in <strong>the</strong> larynx [10, 177]. DCs originatefrom an obstruction <strong>of</strong> <strong>the</strong> gl<strong>and</strong>ular ducts, causedmainly by chronic inflammation. They are mainly locatedon <strong>the</strong> vocal cords, ventricle <strong>of</strong> Morgagni , ventricularfolds , aryepiglottic folds , <strong>and</strong> on <strong>the</strong> pharyngeal side <strong>of</strong><strong>the</strong> epiglottis, where <strong>the</strong>y tend to be larger, even up to7.5 cm in diameter (Fig. 7.1a) [14, 177].The origin <strong>of</strong> a so-called epidermoid cyst <strong>of</strong> <strong>the</strong> vocalcord is probably related to microtraumatic inclusion <strong>of</strong>small fragments <strong>of</strong> squamous epi<strong>the</strong>lium into <strong>the</strong> subepi<strong>the</strong>lialtissue or to <strong>the</strong> remnants <strong>of</strong> <strong>the</strong> vocal cord sulcus[251]. These cysts are usually smaller than o<strong>the</strong>r laryngealretention cysts , measuring 1–4 mm <strong>and</strong> not exceeding10 mm in diameter [80].Laryngoscopically, ductal cysts are seen as a sharplydelineated spherical protrusion, <strong>the</strong> overlying mucosa issmooth <strong>and</strong> stretched. Larger cysts, mainly in newbornsor in small children, can obstruct breathing.The histological picture <strong>of</strong> DCs is influenced by origin.Laryngeal retention cysts are covered in doublelayeredcylindrical, cuboidal or flattened ductal epi<strong>the</strong>lium(Fig. 7.1b) . Squamous or oncocytic metaplasia <strong>of</strong><strong>the</strong> ductal epi<strong>the</strong>lium, partial or complete, is frequentlypresent. Classical “epidermoid” or keratinising cysts<strong>of</strong> <strong>the</strong> vocal cords are usually lined with atrophic keratinisingepi<strong>the</strong>lium with intraluminal stratified basophilickeratin scales . The <strong>the</strong>rapy <strong>of</strong> choice for DC is surgicalremoval.7.2.5 Oncocytic CystAlthough <strong>the</strong> oncocytic lesions dominate in <strong>the</strong> parotidgl<strong>and</strong>, <strong>the</strong>y may appear in <strong>the</strong> minor salivary gl<strong>and</strong>s <strong>of</strong><strong>the</strong> upper aerodigestive tract, including <strong>the</strong> larynx. Awhole spectrum <strong>of</strong> oncocytic laryngeal lesions has beenobserved, ranging from focal to diffuse oncocytic metaplasia,papillary cystic hyperplastic lesions to benign<strong>and</strong> malignant tumours (<strong>the</strong> latter occur mainly in <strong>the</strong>sinonasal <strong>and</strong> palatal region) [44]. On <strong>the</strong> o<strong>the</strong>r h<strong>and</strong>, ithas been suggested that all <strong>the</strong>se lesions, variously calledoncocytic cyst (OC), oncocytic papillary cystadenoma,oncocytoma, oncocytic adenomatous hyperplasia, morelikely belong to non-neoplastic ra<strong>the</strong>r than to true neoplasticlesions [193, 265, 291]. This opinion has beensupported by <strong>the</strong> various extent <strong>of</strong> oncocytic metaplasiain <strong>the</strong> laryngeal minor salivary gl<strong>and</strong>s, as well as by <strong>the</strong>occasional appearance <strong>of</strong> multiple cystic lesions [84, 111,230, 387].Oncocytes are enlarged cells with characteristicgranular eosinophilic cytoplasm, caused by an increasednumber <strong>of</strong> tightly packed abnormal mitochondria,<strong>and</strong> small, dense, darkly stained nuclei. The exactcause <strong>of</strong> oncocytic metaplasia remains unknown,but it is related to <strong>the</strong> process <strong>of</strong> aging <strong>and</strong> especiallyto disturbance <strong>of</strong> <strong>the</strong> organisation <strong>of</strong> <strong>the</strong> mitochondrialenzymes [238]. Laryngeal OCs may show focal,inconspicuous or extensive proliferation <strong>of</strong> oncocytes,mainly with unilocular or multilocular cystic formationswith papillary projections, resembling <strong>the</strong> Warthin’stumour [119].Laryngeal OCs probably represent a separate clinicopathologicentity, showing typical age group, location<strong>and</strong> histopathologic features. They occur on <strong>the</strong> falsevocal cords <strong>and</strong> ventricles in middle-aged to elderly personswith hoarseness or a cough as <strong>the</strong> leading symptoms[216, 265, 283, 304]. Clinically, OCs appear as solitarypolypoid lesions in <strong>the</strong> subepi<strong>the</strong>lial stroma, while


Larynx <strong>and</strong> Hypopharynx Chapter 7 201An outpouching <strong>of</strong> <strong>the</strong> dorsal hypopharyngeal wallabove <strong>the</strong> upper oesophageal sphincter is known asZenker’s diverticulum (ZD). The condition is more <strong>of</strong>tenseen in nor<strong>the</strong>rn Europe, especially <strong>the</strong> UK, thanelsewhere in <strong>the</strong> world [42]. The site <strong>of</strong> origin is between<strong>the</strong> thyropharyngeal <strong>and</strong> <strong>the</strong> more horizontalpart <strong>of</strong> <strong>the</strong> cricopharyngeal muscle. The aetiologicalfactors <strong>of</strong> ZD occurrence have not been explained, butan incomplete sphincter opening with an increase inhypopharyngeal pressure during swallowing has tobe considered [266]. The lesion, which usually occursin elderly persons, is now widely accepted to be <strong>of</strong> acquiredra<strong>the</strong>r than congenital origin. The symptomsare virtually pathognomonic: dysphagia, regurgitation<strong>of</strong> undigested food, weight loss, foetor ex ore, coughing<strong>and</strong> repeated aspiration [42]. Diagnosis <strong>of</strong> <strong>the</strong> diseasemay be confirmed by a barium swallow. Histologically,ZD is composed <strong>of</strong> a squamous epi<strong>the</strong>lium, thinned fibroustissue <strong>of</strong> <strong>the</strong> subepi<strong>the</strong>lial stroma with possibleinflammatory changes. The conservative open surgicalprocedures provide effective results [266]. Exceedinglyrarely, squamous cell carcinoma may develop inZD [42].7.2.7 Aberrant Thyroid TissueFig. 7.2. Oncocytic cyst <strong>of</strong> <strong>the</strong> ventricular cord. Cyst is lined withoncocytic epi<strong>the</strong>liuma diffuse involvement as oncocytic cystadenomatosis isexceptional [230].Histologically, <strong>the</strong> epi<strong>the</strong>lium <strong>of</strong> an OC shows papillaryproliferations or a different degree <strong>of</strong> folding <strong>of</strong><strong>the</strong> cystic wall. The epi<strong>the</strong>lium is typically double layered;<strong>the</strong> inner layer consists <strong>of</strong> columnar eosinophiliccells encircling <strong>the</strong> cystic lumina, while <strong>the</strong> outer layeris composed <strong>of</strong> small basal cells (Fig. 7.2) . Complete endoscopicalsurgical excision is <strong>the</strong> recommended treatment,if necessary by laryng<strong>of</strong>issure [216].7.2.6 Zenker’sHypopharyngeal DiverticleThyroid tissue rarely appears in sites outside <strong>of</strong> itsembryonic development. The subglottic area <strong>of</strong> <strong>the</strong>larynx <strong>and</strong> upper trachea are places where aberrantthyroid tissue (ATT) may be found [36], especiallybetween <strong>the</strong> lower border <strong>of</strong> <strong>the</strong> cricoid cartilage <strong>and</strong><strong>the</strong> upper ring <strong>of</strong> <strong>the</strong> trachea. According to differentreports, intraluminal thyroid tissue occurs anywherebetween <strong>the</strong> glottis <strong>and</strong> <strong>the</strong> bifurcation <strong>of</strong> <strong>the</strong> trachea,as a broad-based, smooth, rounded mass protrudingfrom <strong>the</strong> left subglottic posterolateral wall [305, 345].It has been pointed out that two-thirds <strong>of</strong> patients aremiddle-aged women from regions <strong>of</strong> endemic goitre[22, 36]. Intralaryngotracheal thyroid is a rare lesion.Only about 125 cases were described up to 1998 [327].Waggoner divided intralaryngotracheal thyroid tissueinto “false” <strong>and</strong> “true” aberrant thyroids. The formeris likely to arise in <strong>the</strong> pre- or neonatal period,when <strong>the</strong> thyroid gl<strong>and</strong> could grow into incompletelyformed laryngotracheal cartilages that remain in continuitywith <strong>the</strong> thyroid gl<strong>and</strong>. The latter, <strong>the</strong> “trueaberrant thyroid”, develops during <strong>the</strong> foetal periodas an isolated, misplaced thyroid tissue, when <strong>the</strong> thyroidgl<strong>and</strong> is encroached upon <strong>and</strong> divided by <strong>the</strong> laterdeveloped laryngeal <strong>and</strong> tracheal cartilages [327, 361,372]. The most common symptom <strong>of</strong> intralaryngealATT is slowly progressive dyspnoea, but it may be alsoasymptomatic [278].Histologically, <strong>the</strong> thyroid follicles are usually small,regular, with a well-formed colloid lying close to <strong>the</strong> seromucinousgl<strong>and</strong>s in <strong>the</strong> laryngeal mucosa [238]. Theoverlying mucosa is commonly intact, <strong>and</strong> <strong>the</strong>re may besome evidence <strong>of</strong> chronic irritation. The finding <strong>of</strong> thyroidtissue in <strong>the</strong> laryngotracheal wall raises <strong>the</strong> questionas to whe<strong>the</strong>r or not it represents ectopic tissue appearingthrough a developmental defect or a well-differentiatedcarcinoma. The final decision must be based onan overall clinical evaluation <strong>and</strong> not only on histologicalfindings [36, 327]. Management <strong>of</strong> ATT is <strong>of</strong>ten notclear-cut, but is proposed to be primarily surgical [327,345, 372].


202 N. Gale · A. Cardesa · N. Zidar77.2.8 TracheopathiaOsteochondroplasticaTracheopathia osteochondroplastica (TO) is a rare,slowly progressing lesion, characterised by <strong>the</strong> presence<strong>of</strong> cartilaginous <strong>and</strong> bony submucosal nodules projectinginto <strong>the</strong> lumen <strong>of</strong> <strong>the</strong> trachea, larynx <strong>and</strong> majorbronchi [148, 279, 295, 355]. Most cases are recognisedat autopsy, but may be also suspected due to problems <strong>of</strong>endotracheal intubation [186].The aetiology <strong>and</strong> pathogenesis remain uncertain.Chronic infections, chemical <strong>and</strong> mechanical irritations,metabolic disorders, ecchondrosis, exostosis <strong>and</strong>metaplasia <strong>of</strong> <strong>the</strong> elastic tissue are thought to be causalfactors [279, 295, 355, 356]. TO appears predominantlyin late adult life, but may be seen in childhood <strong>and</strong> earlyadult life [148, 295]. TO with minimal expressions may<strong>of</strong>ten be overlooked. Typical florid cases narrow <strong>the</strong> airways<strong>and</strong> cause a dry cough, dyspnoea, hoarseness <strong>and</strong>recurrent infections [356]. Laryngobronchoscopic examinationis decisive for <strong>the</strong> diagnosis. Elevated, hard,whitish nodules bulge into <strong>the</strong> lumen with <strong>the</strong> appearance<strong>of</strong> a stalactite cave. The posterior wall <strong>of</strong> <strong>the</strong> tracheais usually spared [186, 279]. An intralaryngeal location<strong>of</strong> TO is very rare; as a rule it appears in <strong>the</strong> subglotticregion [355], exceptionally around <strong>the</strong> arytenoids [279].Histologically, submucosal nodules <strong>of</strong> cartilage <strong>and</strong>lamellar bone with marrow spaces are characteristicfindings, usually in relation to <strong>the</strong> underlying cartilage.Calcifications, ossifications <strong>and</strong> fatty marrow formationsmay be seen within <strong>the</strong> nodules [295]. Bone morphogeneticprotein-2 <strong>and</strong> transforming growth factorbeta, <strong>the</strong> potent inducers <strong>of</strong> new bone formation, havebeen recently detected in TO immunohistochemically<strong>and</strong> may have some decisive roles in <strong>the</strong> pathogenesis[356]. Surgical treatment has been recommended onlyin symptomatic patients [35, 186, 250].7.3 Inflammatory Lesions7.3.1 Acute Infections7.3.1.1 EpiglottitisAcute epiglottitis (AE), or more precisely termed supraglottitis[121, 331], is a potential risk <strong>of</strong> fatal airway obstructionin previously healthy persons. In <strong>the</strong> past, AEwas mainly a childhood disease caused by Haemophilusinfluenzae type B. Due to <strong>the</strong> introduction <strong>of</strong> an immunisationprogramme in <strong>the</strong> late 1980s, <strong>the</strong> diseasehas been steadily decreasing in children, but still has ahigh incidence in <strong>the</strong> adult population, more frequentlyin a form related to infections with pyogenic cocci [75,87, 231]. The most consistently found presenting symptomis severe pain on swallowing. In children, breathingdifficulties are <strong>of</strong>ten <strong>the</strong> predominant symptom.O<strong>the</strong>r symptoms <strong>and</strong> signs are hoarseness, drooling, fever,tachycardia <strong>and</strong> toxic appearance [75, 87]. Reddish<strong>and</strong> evidently oedematous supraglottic area, includingtongue <strong>and</strong> pharyngeal structures, is observed. Oedematousswelling rarely spreads to <strong>the</strong> glottic region.Microscopic examination shows diffuse exudative inflammationwith fibrin, neutrophils <strong>and</strong> erythrocytesinvolving supraglottic structures. An early complaint <strong>of</strong>dyspnoea may safely discriminate between patients requiringinvasive airway management with intubation <strong>and</strong>conservative treatment with close observation [150].7.3.1.2 LaryngotracheobronchitisLaryngotracheobronchitis (LTB), also known as subglotticlaryngitis, non-diph<strong>the</strong>ric croup, virus croup,spasmodic croup <strong>and</strong> fibrinous LTB, <strong>of</strong>ten occurs inchildren aged 1–3 years. Generally, LTB is <strong>of</strong> limitedduration, caused by influenza, parainfluenza or o<strong>the</strong>rviruses. Prolonged infection by o<strong>the</strong>r pathogens maybe also involved [170]. The onset <strong>of</strong> <strong>the</strong> disease is moregradual compared with acute epiglottitis. When fullydeveloped, a croupy cough with inspiratory <strong>and</strong> expiratorystridor is present. Histologically, characteristicfibrinous laryngitis is observed with destruction <strong>of</strong> <strong>the</strong>respiratory epi<strong>the</strong>lium. The mortality rate <strong>of</strong> <strong>the</strong> diseasehas remained low for many years [238, 353].7.3.1.3 Diph<strong>the</strong>riaFortunately, laryngeal diph<strong>the</strong>ria is a matter <strong>of</strong> historynow in <strong>the</strong> developed world. Very exceptionally, an individualcase has been reported [137]. Histologically,dirty white, fibrinosuppurative membranes covering<strong>the</strong> laryngeal mucosa, accompanied by a foul smell, arecharacteristic <strong>of</strong> <strong>the</strong> disease [203].7.3.2 Chronic Infections7.3.2.1 TuberculosisLaryngeal tuberculosis (LT) was considered one <strong>of</strong> <strong>the</strong>most common diseases in <strong>the</strong> pre-antibiotic period, affecting<strong>the</strong> larynx in 35 to 83% <strong>of</strong> patients with pulmonarytuberculosis [268, 390]. By <strong>the</strong> 1980s, <strong>the</strong> diseasehad become very rare in <strong>the</strong> developed world, owing to<strong>the</strong> advent <strong>of</strong> antibiotic <strong>the</strong>rapy, immunisation <strong>and</strong> improvedsocial <strong>and</strong> economic conditions. However, since1980, tuberculosis has again been showing a rising in-


Larynx <strong>and</strong> Hypopharynx Chapter 7 203cidence worldwide, including developed countries,owing to <strong>the</strong> spread <strong>of</strong> <strong>the</strong> HIV infection, poor livingst<strong>and</strong>ards with malnutrition, <strong>the</strong> emergence <strong>of</strong> drugresistantmycobacteria <strong>and</strong> immigration from countrieswhere tuberculosis is still endemic [61, 78, 125,184, 268, 311, 315]. Consequently, <strong>the</strong> World HealthOrganisation has declared tuberculosis to be a globalemergency [258].LT currently affects mostly males; <strong>the</strong> average age<strong>of</strong> patients is about 50, with a history <strong>of</strong> heavy drinking<strong>and</strong> smoking. The most common presenting symptomis dysphonia, followed by dysphagia, odynophagia,stridor, a cough, <strong>and</strong> haemoptysis, generally associatedwith more or less obvious signs <strong>of</strong> pulmonary involvement[315, 390]. The true vocal cords are most commonlyaffected, although <strong>the</strong> supraglottic region is also involved[311]. The majority <strong>of</strong> cases present as hypertrophic,exophytic, hyperaemic lesions, sometimes nodularor ulcerated.Histologically, <strong>the</strong> subepi<strong>the</strong>lial stroma contains caseatinggranulomas with a central caseous necrosis, surroundedby epi<strong>the</strong>lioid macrophages, Langerhans-typegiant cells <strong>and</strong> lymphocytes (Fig. 7.3) . The covering epi<strong>the</strong>liummay be normal, ulcerated or show pseudoepi<strong>the</strong>liomatoushyperplasia. Identification <strong>of</strong> Mycobacteriumtuberculosis by special stainings or molecular geneticmethods confirms <strong>the</strong> diagnosis <strong>of</strong> LB.Differential diagnosis includes a large spectrum <strong>of</strong>granulomatous diseases, such as sarcoidosis, cat-scratchdisease, fungal infections, Wegener’s granulomatosis<strong>and</strong> tumourous lesions. Differentiation between sarcoidosis<strong>and</strong> tuberculosis is difficult. Generally, granulomasin sarcoidosis lack caseation <strong>and</strong> stainings formycobacteria are negative. Cat-scratch disease can beruled out by <strong>the</strong> presence <strong>of</strong> rounded or stellate granulomascontaining central granular debris <strong>and</strong> neutrophils.Fungal granulomas can be confirmed by identification<strong>of</strong> <strong>the</strong> microorganism. Granulomas in Wegener’sgranulomatosis are not closely packed, fibrinoid necrosis<strong>of</strong> collagen is prominent <strong>and</strong> vasculitis is occasionallypresent.The treatment <strong>of</strong> LT primarily consists <strong>of</strong> antituberculoustreatment, while surgical procedure is reservedfor cases <strong>of</strong> air compromise [390].7.3.2.2 Fungal InfectionsFig. 7.3. Laryngeal tuberculosis. Granulomas are composed <strong>of</strong>epi<strong>the</strong>lioid cells, Langerhans giant cells without necrosis, surroundedby mononuclear inflammatory cellsFungal infections <strong>of</strong> <strong>the</strong> larynx are very rare, but may beexpected to arise, though not exclusively, in immunocompromisedpatients. Mycotic infections tend to occuras a result <strong>of</strong> <strong>the</strong> dissemination <strong>of</strong> <strong>the</strong> fungi, especiallyfrom <strong>the</strong> bronchopulmonary foci. An important exceptionis laryngeal c<strong>and</strong>idiasis, which usually occurs as aresult <strong>of</strong> direct spread from <strong>the</strong> oral cavity [280]. Differenttypes <strong>of</strong> mycotic infections have been reported,such as laryngeal histoplasmosis [302, 319], cryptococcosis[171, 236], coccidioidomycosis [41], blastomycosis[89, 307], c<strong>and</strong>idiasis [280], paracoccidioidomycosis[317] <strong>and</strong> aspergillosis [272, 310]. The histological featuresare similar for each <strong>of</strong> <strong>the</strong>se infections <strong>and</strong> rangefrom granulomatous lesions related to histoplasma <strong>and</strong>cryptoccocus, to abscess formation in blastomycosis<strong>and</strong> aspergillosis [291]. Pronounced epi<strong>the</strong>lial hyperplasiawith prominent ortho-parakeratosis or pseudoepi<strong>the</strong>liomatoushyperplasia in laryngeal blastomycosis,c<strong>and</strong>idiasis <strong>and</strong> aspergillosis may mimic <strong>the</strong> squamouscell <strong>and</strong> verrucous cell carcinoma [272, 280, 307]. Theidentification <strong>of</strong> <strong>the</strong> causal agents by special silver, PASor mucicarmine stains <strong>of</strong> <strong>the</strong> biopsy specimens <strong>and</strong>/orcultures <strong>of</strong> microorganisms are crucial for successfultreatment.7.3.2.3 O<strong>the</strong>r Rare InfectionsIn non-European countries, <strong>the</strong> larynx is occasionallyinvolved in rare infections, such as rhinoscleroma [7,100, 275], leprosy [145, 342], leishmaniasis <strong>and</strong> trichinosis[238].7.3.3 Non-InfectiousInflammatory Lesions7.3.3.1 Wegener’s GranulomatosisWegener’s granulomatosis (WG) is a systemic diseasecharacterised by necrotising vasculitis, formation <strong>of</strong>granulomas in <strong>the</strong> upper <strong>and</strong> lower respiratory tracts,


204 N. Gale · A. Cardesa · N. Zidar7<strong>and</strong> glomerulonephritis. Limited forms <strong>of</strong> WG also occur,<strong>of</strong>ten with involvement <strong>of</strong> <strong>the</strong> respiratory tract,but without kidney involvement. The vast majority <strong>of</strong>patients have antineutrophil cytoplasmic antibodies(ANCA) in <strong>the</strong> serum with a characteristic cytoplasmicpattern (C-ANCA) [172].The upper respiratory tract is <strong>the</strong> commonest presentingsite <strong>of</strong> WG, mainly affecting <strong>the</strong> paranasal sinuses,followed by <strong>the</strong> nose, nasopharynx <strong>and</strong> larynx[82]. Local clinical symptoms <strong>and</strong> signs, such as rhinorrhoea,pain, mucosal ulcerations <strong>and</strong> hoarseness,are non-specific. They may be accompanied by systemicsymptoms <strong>and</strong> signs, such as fever, weakness <strong>and</strong>weight loss [339].Histological features include inflammation, necrotisinggranulomas, <strong>and</strong> vasculitis. Necrosis in WG hasa patchy distribution, with serpiginous borders, <strong>and</strong> isusually basophilic, with a finely granular appearance.Granulomas tend to be loose, not closely packed as insarcoidosis or tuberculosis [82]. Vasculitis typically involvessmall to medium-sized arteries <strong>and</strong> veins, withany <strong>of</strong> <strong>the</strong> following features: fibrinoid necrosis, fragmentation<strong>of</strong> <strong>the</strong> elastic lamina, acute <strong>and</strong> chronic inflammatorycells <strong>and</strong> granulomas. The lesions may undergoorganisation <strong>and</strong> fibrosis.The diagnosis <strong>of</strong> WG is based on clinical features, biopsy<strong>of</strong> <strong>the</strong> related lesions, <strong>and</strong> <strong>the</strong> cytoplasmic pattern<strong>of</strong> anti-neutrophil cytoplasmic antibodies (C-ANCA) in<strong>the</strong> serum [102]. A positive biopsy <strong>of</strong> <strong>the</strong> upper respiratorytract has a high predictive value, up to 100%, indicatingfew or even no false-positive results [173]. However,vasculitis is only rarely seen on biopsy. Histology<strong>of</strong>ten reveals non-specific features – inflammation <strong>and</strong>necrosis, with or without granuloma formation [397].Wegener’s granulomatosis should be differentiatedfrom o<strong>the</strong>r forms <strong>of</strong> vasculitis, o<strong>the</strong>r granulomatous diseases,cocaine abuse, <strong>and</strong> from neoplasms, particularlyNK/T lymphoma <strong>of</strong> <strong>the</strong> nasal type [339]. The presence<strong>of</strong> C-ANCA proves extremely helpful in differentiationfrom almost all <strong>the</strong> diseases mentioned [173].Wegener’s granulomatosis was almost universally fatalin <strong>the</strong> past, usually within a few months <strong>of</strong> <strong>the</strong> onset<strong>of</strong> clinically apparent renal disease. However, with modernimmunosuppressive <strong>the</strong>rapy, <strong>the</strong> prognosis <strong>of</strong> WG isexcellent. A marked improvement is seen in 90% <strong>of</strong> patients<strong>and</strong> complete remission is achieved in 75% <strong>of</strong> patients[160]. Early detection <strong>of</strong> WG is essential to preventfully developed disease. For WG at o<strong>the</strong>r sites in <strong>the</strong>head <strong>and</strong> neck, see also Chaps. 2 <strong>and</strong> 3.7.3.3.2 SarcoidosisSarcoidosis is a chronic granulomatous disease <strong>of</strong> unknownaetiology that can affect any organ system. Inaddition to <strong>the</strong> classic involvement <strong>of</strong> lungs, hilar <strong>and</strong>mediastinal lymph nodes, <strong>the</strong> eyes, skin, liver, bones<strong>and</strong> nervous system may also be affected. Laryngeal involvementis usually a part <strong>of</strong> generalised disease withan incidence <strong>of</strong> 1–5% [72, 197]. However, laryngeal sarcoidosiscan also appear as an isolated disease [40, 197,263]. The supraglottic region is mostly affected, especially<strong>the</strong> epiglottis, aryepiglottic folds <strong>and</strong> arytenoids,showing oedematous, pale, diffusely enlarged mucosawith occasional nodularity, which has been considered<strong>the</strong> pathognomonic feature <strong>of</strong> laryngeal disease [29, 72,263]. Subglottic <strong>and</strong> true vocal cord involvement is rare[40, 237, 263]. The disease is usually self-healing withan inconspicuous course including remissions <strong>and</strong> exacerbations.Histologically, non-caseating <strong>and</strong> non-confluentgranulomas are a characteristic feature. Granulomas arecomposed <strong>of</strong> epi<strong>the</strong>lioid cells <strong>and</strong> Langerhans-type giantcells with no central necrosis. Two structures are <strong>of</strong>tenfound in giant cells, although <strong>the</strong>y are not pathognomonicfor sarcoidosis: asteroid bodies are stellate crystallineinclusions, <strong>and</strong> laminated concretions, composed<strong>of</strong> calcium <strong>and</strong> proteins, known as Schaumann bodies.The sarcoid granulomas can be transformed into hyalinefibrous scars.In histologically proven non-caseating granulomas,o<strong>the</strong>r laryngeal granulomatous diseases, such as infectiousgranulomatous diseases, granulomatous processes<strong>of</strong> unknown pathogenesis (Wegener’s granulomatosis)<strong>and</strong> inhalant granulomatous processes (berylliosis,asbestosis), must be excluded [189]. No microorganismsare found in sarcoidal granulomas with some exceptions,such as cell wall-deficient forms <strong>of</strong> mycobacteria[294]. In contrast to tuberculosis <strong>and</strong> histoplasmosis,sarcoidal granulomas are non-caseating. Sarcoidosislacks vasculitis, characteristic <strong>of</strong> Wegener’s granulomatosis.Inhalant granulomatous diseases, which result insignificant pulmonary fibrosis, rarely affect <strong>the</strong> laryngealmucosa [189].Early diagnosis <strong>and</strong> adequate treatment <strong>of</strong> laryngealsarcoidosis is important to prevent upper airway obstruction<strong>and</strong> tracheotomy. Although <strong>the</strong> course <strong>of</strong> diseasemay be <strong>of</strong> long duration, spontaneous remissionsusually occur. When treatment is necessary, <strong>the</strong> administration<strong>of</strong> intralesional or systemic steroids is performed[72].7.3.3.3 Rheumatoid ArthritisRheumatoid arthritis (RA) is a chronic systemic, presumablyautoimmune disorder, characterised by proliferativesynovitis that <strong>of</strong>ten progresses to destruction<strong>of</strong> <strong>the</strong> articular cartilage <strong>and</strong> ankylosis <strong>of</strong> <strong>the</strong> joints.O<strong>the</strong>r tissues <strong>and</strong> organs may also be affected, suchas <strong>the</strong> skin, blood vessels, heart, lung, nervous system,etc.


Larynx <strong>and</strong> Hypopharynx Chapter 7 205Laryngeal involvement in RA includes arthritis <strong>of</strong> <strong>the</strong>cricoarytenoid <strong>and</strong> cricothyroid joints, <strong>and</strong>/or <strong>the</strong> formation<strong>of</strong> rheumatoid nodules in <strong>the</strong> s<strong>of</strong>t tissue <strong>of</strong> <strong>the</strong>larynx [47, 95, 130].Histologically, <strong>the</strong> acute phase <strong>of</strong> arthritis is characterisedby swelling <strong>and</strong> thickening <strong>of</strong> <strong>the</strong> synovia, whichis heavily infiltrated by mononuclear cells, resulting invillous hypertrophy. In <strong>the</strong> chronic phase, <strong>the</strong>re is destruction<strong>of</strong> <strong>the</strong> articular cartilage <strong>and</strong> proliferation <strong>of</strong>fibrous tissue with obliteration <strong>of</strong> <strong>the</strong> joint spaces, rarelyleading to bony ankylosis [47].Rheumatoid nodules may develop in <strong>the</strong> s<strong>of</strong>t tissueadjacent to <strong>the</strong> joints, or in <strong>the</strong> vocal cords. Microscopically,<strong>the</strong>y consist <strong>of</strong> a central fibrinoid necrosis, surroundedby palisading macrophages, lymphocytes <strong>and</strong>plasma cells (Fig. 7.4) [95, 343].In <strong>the</strong> acute phase symptoms <strong>and</strong> signs are usuallymild, <strong>and</strong> consist <strong>of</strong> pain <strong>and</strong> voice disturbance. In <strong>the</strong>chronic phase, dyspnoea <strong>and</strong> respiratory obstructionmay develop [2, 39, 95].Arthritis <strong>and</strong> rheumatoid nodules are not pathognomonic<strong>of</strong> RA, but are also seen in patients with o<strong>the</strong>r autoimmunediseases, particularly systemic lupus ery<strong>the</strong>matosus[241, 366].Rheumatoid arthritis is treated by anti-inflammatorydrugs <strong>and</strong> local administration <strong>of</strong> steroids by means<strong>of</strong> injection or aerosol; in rare cases, surgical <strong>the</strong>rapy isneeded to relieve airway obstruction [2, 27, 39, 95].7.3.3.4 Relapsing PolychondritisRelapsing polychondritis (RP) is a rare autoimmunedisease characterised by progressive inflammation<strong>of</strong> cartilaginous structures, both elastic <strong>and</strong> hyaline,throughout <strong>the</strong> body [68, 233, 364]. The disease typicallyinvolves <strong>the</strong> ear, nose, eye, larynx <strong>and</strong> lower respiratorytract, costal cartilages, joints, cardiovascularsystem, renal tissue <strong>and</strong> central nervous system. Themost serious consequence <strong>of</strong> RP in <strong>the</strong> larynx <strong>and</strong> tracheais destruction <strong>of</strong> <strong>the</strong> cartilaginous framework,with collapse <strong>of</strong> <strong>the</strong> airway <strong>and</strong> breathing difficulties.RP commonly affects patients between 40 <strong>and</strong> 60 years[208], <strong>and</strong> a female-to-male ratio <strong>of</strong> 3:1 was recentlynoted [364]. Aetiologically, <strong>the</strong>re is strong indication <strong>of</strong>an autoimmune origin <strong>of</strong> RP [364], which has recentlybeen supported by <strong>the</strong> finding <strong>of</strong> antibodies to type IIcollagen in two-thirds <strong>of</strong> patients [349], <strong>and</strong> linkage to<strong>the</strong> human leukocyte antigen DR4 gene <strong>and</strong> o<strong>the</strong>r autoimmunediseases. The onset <strong>of</strong> RP is generally sudden,with recurrent attacks <strong>of</strong> acute inflammation <strong>of</strong><strong>the</strong> auricle. The diagnosis <strong>of</strong> RP is believed to be convincingif patients have at least three <strong>of</strong> <strong>the</strong> followinginvolvements: bilateral auricular lesions, seronegativearthritis, nasal <strong>and</strong> ocular affections, respiratory tractFig. 7.4. Rheumatoid nodule <strong>of</strong> <strong>the</strong> vocal cord. Fibrinoid necrosissurrounded by palisading macrophages, lymphocytes <strong>and</strong> plasmacellschondritis, audiovestibular damages <strong>and</strong> histologicalconfirmation <strong>of</strong> <strong>the</strong> disease [233].Hoarseness, coughing, dyspnoea, choking <strong>and</strong> tendernessover <strong>the</strong> laryngotracheal cartilages are symptoms<strong>of</strong> laryngeal <strong>and</strong> lower respiratory tract involvement[152]. Although airway obstruction could be localisedto <strong>the</strong> glottic <strong>and</strong> subglottic area, a diffuse involvement<strong>of</strong> <strong>the</strong> respiratory tract is more common [247]<strong>and</strong> occurs in up to 50% <strong>of</strong> patients with RP [93].Histologically, <strong>the</strong> affected cartilage shows distinctfeatures: <strong>the</strong> cartilaginous matrix loses its basophilicstaining, peri-<strong>and</strong> intracartilaginous inflammation isevident, inflammatory cells infiltrate <strong>the</strong> perichondrium<strong>and</strong> cartilage. The chondrocytes become vacuolated<strong>and</strong> necrotic, <strong>and</strong> fragmentation <strong>of</strong> cartilage is evident.With <strong>the</strong> progression <strong>of</strong> <strong>the</strong> disease, necrotic cartilageis replaced by granulation tissue <strong>and</strong> later by fibrosis.In <strong>the</strong> course <strong>of</strong> <strong>the</strong> disease, persistent inflammationcan destroy <strong>the</strong> cartilaginous rings <strong>and</strong> cause luminalcollapse.Differential diagnosis includes all conditions that essentiallyshow cartilage destruction: various infectious(tuberculosis <strong>and</strong> o<strong>the</strong>r bacterial, fungal, <strong>and</strong> viral infections),non-infectious diseases (sarcoidosis, Wegener’sgranulomatosis <strong>and</strong> o<strong>the</strong>r types <strong>of</strong> systemic vasculitides)<strong>and</strong> tumours (lymphoma, cartilaginous tumours).Treatment for RP is based on systemic steroid <strong>and</strong> occasionallyimmunosuppressive <strong>the</strong>rapy [261]. Involvement<strong>of</strong> <strong>the</strong> larynx <strong>and</strong> <strong>the</strong> lower respiratory tract is <strong>the</strong>most serious complication. The mortality rate reported


206 N. Gale · A. Cardesa · N. Zidar7in 1972 was 50% [164], <strong>and</strong> had fallen to 10% 14 yearslater [239]. Early diagnosis can influence a better outcome,<strong>and</strong> survival rate appears more favourable thanwas previously thought [364].7.3.3.5 GoutGout is a disorder <strong>of</strong> <strong>the</strong> purine metabolism typicallyidentified by hyperuricaemia. The disease is characterisedby temporary attacks <strong>of</strong> acute arthritis caused byprecipitation <strong>of</strong> monosodium urate crystals within <strong>and</strong>about <strong>the</strong> joints. The course <strong>of</strong> <strong>the</strong> disease may eventuallylead to deposition <strong>of</strong> urates in <strong>the</strong> joints <strong>and</strong> o<strong>the</strong>rs<strong>of</strong>t tissues, creating tophi. The first attack <strong>of</strong> <strong>the</strong> diseaseis mainly monoarticular with a predilection to <strong>the</strong> firstmetatarsophalangeal joint. The head <strong>and</strong> neck regionis rarely involved, although <strong>the</strong> classic site is <strong>the</strong> externalear. In addition, tophi may appear in <strong>the</strong> intervertebraldiscs, oropharynx, temporom<strong>and</strong>ibular joint,<strong>and</strong> tongue. There has been limited evidence <strong>of</strong> chronicgouty involvement <strong>of</strong> <strong>the</strong> larynx, with less than 20 caseshaving been reported so far [365]. Acute gouty cricoary<strong>the</strong>noidarthritis is <strong>the</strong> most common <strong>and</strong> may giverise to pain within <strong>the</strong> larynx, dysphonia, odynophagia,dysphagia or stridor. After repeated attacks, <strong>the</strong> articularcartilage is gradually destroyed leading to ankylosis<strong>of</strong> <strong>the</strong> joint. The fixed vocal cord may mimic growth <strong>of</strong>a malignant tumour [365]. Tophi <strong>of</strong> <strong>the</strong> laryngeal s<strong>of</strong>ttissue are exceedingly rare [146], <strong>the</strong> involved mucosa<strong>of</strong> <strong>the</strong> vocal cords shows a granular surface [228]. Thehistological features <strong>of</strong> tophi are conspicuous, with largeaggregates <strong>of</strong> needle-shaped urate crystals (birefringentcrystalline deposits) surrounded by macrophages, foreign-typegiant cells, lymphocytes <strong>and</strong> fibroblasts [146].In more remote differential diagnostic possibilities, o<strong>the</strong>rlesions with deposition <strong>of</strong> various substances in <strong>the</strong>laryngeal mucosa, such as amyloid or Teflon, need to beconsidered.7.3.3.6 Teflon GranulomaInjection <strong>of</strong> Teflon paste (polytetrafluoroethylene)into <strong>the</strong> lateral thyreoarytenoid muscle has been usedin patients with unilateral paralysis <strong>of</strong> <strong>the</strong> vocal cord,with <strong>the</strong> aim <strong>of</strong> augmenting <strong>and</strong> medialising <strong>the</strong> paralysedhemilarynx. The increased bulk <strong>of</strong> <strong>the</strong> vocal cordcould, <strong>the</strong>refore, contribute to a more complete glotticclosure, prevent aspiration, <strong>and</strong> improve a poor-qualityvoice caused by impaired breathing. The most commoncause <strong>of</strong> vocal cord paralysis is surgical trauma <strong>of</strong> <strong>the</strong>laryngeal recurrent nerve <strong>and</strong>/or malignant tumours[31, 370, 385]. In general, Teflon injection has been welltolerated, <strong>and</strong> after a short-lived inflammatory reaction,it becomes stable <strong>and</strong> walled-<strong>of</strong>f by surrounding fibrosis[31, 210]. Technical errors during injections, such as overinjection or misplaced injection <strong>of</strong> Teflon may, however,cause dysphonia <strong>and</strong> airway obstruction, as well as <strong>the</strong>presence <strong>of</strong> a neck mass, resembling a neoplasm, in <strong>the</strong>case <strong>of</strong> an escape <strong>of</strong> Teflon via <strong>the</strong> cricothyroid membrane[264, 370].Teflon granulomas (TGs) are submucosal polypoidlesions <strong>of</strong> <strong>the</strong> vocal cord. Histologically, <strong>the</strong>y are composed<strong>of</strong> a foreign body giant cell reaction with extensionto <strong>the</strong> underlying muscle <strong>and</strong> cartilage. Teflon ispresent in foreign body giant cells, <strong>and</strong> also extracellularly,as glassy crystalline deposits that are characteristicallybirefringent under polarised light. A dense fibrotictissue is evident over time, while <strong>the</strong> surrounding inflammatoryinfiltrate is not present [385]. TG is treatedby conservative surgery, although <strong>the</strong> results are unpredictable[31]. There have been no reports <strong>of</strong> cancer developmentin TG [385]. With <strong>the</strong> introduction <strong>of</strong> laryngealframework surgery <strong>and</strong> medialisation laryngoplasty,fewer centres nowadays additionally advocate Tefloninjection [370].7.3.3.7 Idiopathic SubglotticLaryngeal StenosisIdiopathic subglottic stenosis (ISS) is a rare, slowlyprogressive inflammatory disease <strong>of</strong> unknown aetiologyinvolving mainly <strong>the</strong> region <strong>of</strong> cricoid cartilage <strong>and</strong><strong>the</strong> first tracheal ring. The pathogenesis <strong>of</strong> <strong>the</strong> diseaseremains hypo<strong>the</strong>tical. ISS has recently been associatedwith various possible causes, such as gastro-oesophagealreflux, autoimmune diseases <strong>and</strong> previous infections<strong>of</strong> <strong>the</strong> respiratory tract [30, 174, 369]. Maronian<strong>and</strong> co-workers suggested that <strong>the</strong> term ISS shouldeven be replaced by reflux-induced subglottic stenosis,if <strong>the</strong>re is no o<strong>the</strong>r clear cause <strong>of</strong> <strong>the</strong> disease [229]. ISShas a strong female predilection [73, 132, 369]. The age<strong>of</strong> females when <strong>the</strong> symptoms start to appear rangesfrom 15 to 75 years (average 43.5 years) [73].The diagnosis <strong>of</strong> disease is a matter <strong>of</strong> exclusion, <strong>and</strong>all o<strong>the</strong>r possible causes <strong>of</strong> a subglottic stenosis mustfirst be ruled out. Aetiologically, subglottic stenoses aremost commonly linked to endolaryngeal trauma, especiallyafter prolonged intubation. O<strong>the</strong>r diseases, includinginfections, laryngotracheal localisation <strong>of</strong> systemicdiseases such as Wegener’s granulomatosis <strong>and</strong>o<strong>the</strong>r collagen vascular diseases, amyloidosis <strong>and</strong> sarcoidosis,are rarely associated with laryngeal stenosis.Various benign <strong>and</strong> malignant tumours may also resembleISS [132, 281]. ISS usually presents as dyspnoea,a cough <strong>and</strong> dysphonia [30, 73, 369].Histological examination characteristically showsa spreading <strong>of</strong> dense fibrous tissue, extending up to<strong>the</strong> surface <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium. Fibrosis is usually poorlycellular, with prominent augmentation <strong>of</strong> thick col-


Larynx <strong>and</strong> Hypopharynx Chapter 7 207lagenous fibres. Some inconspicuous chronic inflammatoryinfiltrate may be present around blood vessels,without evidence <strong>of</strong> vasculitis. The covering epi<strong>the</strong>lium,squamous or respiratory, may be reactively hyperplastic.ISS is a chronic lifetime disease that requiresmultiple surgical dilatations for palliation [73]. Evaluationfor laryngopharyngeal reflux disease should beperformed with pharyngeal pH testing in all patients,in an attempt to clarify <strong>the</strong> aetiology <strong>of</strong> ISS [229]. Moresevere cases are managed with laryngotracheal resection<strong>and</strong> reconstruction [369].7.3.3.8 Angioneurotic OedemaAngioneurotic oedema (ANO) is a rapidly appearing,recurrent, non-pitting oedema <strong>of</strong> <strong>the</strong> subcutaneous<strong>and</strong>/or submucosal tissues causing a life-threateningcondition, affecting <strong>the</strong> larynx, hypo-<strong>and</strong> oropharynx<strong>and</strong> oral cavity [144]. ANO can occur as a result <strong>of</strong> hereditary<strong>and</strong> acquired deficiencies in <strong>the</strong> immune <strong>and</strong>non-immune responses [81]. Several forms <strong>of</strong> ANO arerecognised:1. IgE-dependent, caused by pollens, foods, drugs, fungi,cold, sun <strong>and</strong> exercise;2. Complement-mediated, hereditary <strong>and</strong> acquired withdeficiency <strong>of</strong> <strong>the</strong> C1 esterase inhibitor <strong>of</strong> <strong>the</strong> complementcascade;3. Non-immunologic, direct mast cell-releasing agentscaused by different drugs <strong>and</strong> aspirin, <strong>and</strong> o<strong>the</strong>r nonsteroidalanti-inflammatory drugs that alter <strong>the</strong> arachidonicacid metabolism;4. Idiopathic [13].Angioneurotic oedema, ei<strong>the</strong>r acquired or hereditary,is characterised by sudden onset with full developmentwithin a few hours <strong>and</strong> fades over <strong>the</strong> course<strong>of</strong> 48–72 h. Gastrointestinal mucosa may also be affected,mainly in <strong>the</strong> hereditary disease, causingsevere abdominal pain, nausea, vomiting <strong>and</strong> diarrhoea.Various degrees <strong>of</strong> laryngeal oedema may bepresent, affecting mainly <strong>the</strong> anterior surface <strong>of</strong> <strong>the</strong>epiglottis, aryepiglottic folds, base <strong>of</strong> <strong>the</strong> tongue <strong>and</strong>hypopharynx [238]. Generally, ANO resolves withoutharm, but laryngeal <strong>and</strong> tracheal oedema may causeasphyxiation <strong>and</strong> remains a considerable cause <strong>of</strong>death [90, 362]. The frequency <strong>of</strong> attacks in hereditaryforms varies considerably from less than 1 to 25per year. Lesions can be solitary or multiple, <strong>and</strong> primarilyinvolve <strong>the</strong> extremities, larynx, face <strong>and</strong> bowelwall [90]. Emergency treatment is required if <strong>the</strong> processleads to respiratory distress because <strong>of</strong> laryngealinvolvement.7.4 Degenerative Lesions7.4.1 OculopharyngealMuscular DystrophyOculopharyngeal muscular dystrophy (OPMD) is a lateonset,dominantly inherited, slowly progressing disease,carried by a limited expansion <strong>of</strong> <strong>the</strong> triplet <strong>of</strong> GCGnucleotides in <strong>the</strong> PABP2 gene on chromosome 14q11[43]. Although OPMD has a world-wide distribution,its prevalence is highest in patients <strong>of</strong> French-Canadianorigin [129, 155]. The onset <strong>of</strong> disease occurs in middlelife, most <strong>of</strong>ten presenting with ptosis <strong>and</strong> a slight degree<strong>of</strong> ophthalmoplegia, followed later by dysphagia <strong>and</strong><strong>of</strong>ten proximal limb weakness. The disease progressesslowly, but <strong>the</strong> dysphagia may be severe <strong>and</strong> has beenreported to be a cause <strong>of</strong> death by starvation in severalcases [316]. Muscle biopsy reveals various changes inmuscle fibres, such as atrophy <strong>and</strong> regeneration with anincreased number <strong>of</strong> myocyte nuclei <strong>and</strong> <strong>the</strong>ir centripetalorientation. Some findings, such as intracytoplasmicrimmed vacuoles, found by light microscopy, <strong>and</strong>intranuclear filament inclusions, seen by electron microscopy,are its pathological hallmarks [316]. Hill <strong>and</strong>co-workers provided confirmation that <strong>the</strong> detection <strong>of</strong>exp<strong>and</strong>ed GCG-repeated lengths in <strong>the</strong> PABP2 gene is areliable diagnostic test for OPMD in <strong>the</strong> English population[155]. However, <strong>the</strong>ir results were not in accordancewith o<strong>the</strong>r molecular studies [43]. Simple proceduressuch as blepharoplasty <strong>and</strong> cricopharyngeal myotomyconsiderably improve <strong>the</strong> quality <strong>of</strong> life <strong>of</strong> <strong>the</strong>se patients[129].7.5 Pseudotumours7.5.1 Exudative Lesions<strong>of</strong> Reinke’s SpaceThe special anatomic framework <strong>of</strong> Reinke’s space isconsidered essential for <strong>the</strong> development <strong>of</strong> a group <strong>of</strong>so-called exudative benign lesions <strong>of</strong> <strong>the</strong> vocal cords,including Reinke’s oedema (RO), vocal cord polyps(VCPs) <strong>and</strong> nodules (VCNs) [177, 179, 181, 238, 308].Each <strong>of</strong> three entities has its own clinical <strong>and</strong> morphologicalspecificities [85], but most <strong>of</strong> <strong>the</strong>m overlap. Thecommon basic pathogenetic mechanism is blood vesselinjuries with accumulation <strong>of</strong> oedematous fluid in Reinke’sspace [85, 308].


208 N. Gale · A. Cardesa · N. Zidar7abFig. 7.5. Reinke’s oedema. a Diffuse oedematous swelling in <strong>the</strong>entire length <strong>of</strong> both vocal cords. b Diffusely oedematous, bluecolouredsubepi<strong>the</strong>lial stroma is lined with hyperplastic squamousepi<strong>the</strong>lium with thickened basement membrane. Epi<strong>the</strong>liumshows hyperplasia <strong>of</strong> basal <strong>and</strong> parabasal cells7.5.1.1 Reinke’s OedemaReinke’s oedema is a chronic, diffuse, mainly bilateral,oedematous swelling <strong>of</strong> <strong>the</strong> membranous part <strong>of</strong> <strong>the</strong>vocal cords [177]. Several synonyms for RO have beenused, such as polypoid vocal fold, polypoid degeneration,chronic polypoid chorditis <strong>and</strong> chronic oedematous hypertrophy[320]. The specific morphologic features <strong>of</strong>Reinke’s space , such as sparse lymphatic drainage <strong>and</strong>its sharply demarcated borders, except <strong>the</strong> lateral one,contribute to <strong>the</strong> development <strong>of</strong> RO [158, 177, 179, 308,309]. Various mechanical <strong>and</strong> chemical aetiologic factorsare related to <strong>the</strong> development <strong>of</strong> RO , including overuseor abuse <strong>of</strong> <strong>the</strong> voice, <strong>and</strong> cigarette smoking . The role <strong>of</strong>constitutional <strong>and</strong> hormonal disturbances such as hypothyroidism,remains uncertain [32, 177, 308, 395]. Thelesion appears most commonly in women <strong>of</strong> 20–40 years<strong>of</strong> age with hoarseness as <strong>the</strong> leading symptom.Laryngoscopically, <strong>the</strong> surface <strong>of</strong> <strong>the</strong> swollen vocalcords along <strong>the</strong>ir entire length is smooth, translucent<strong>and</strong> jelly-like, with a clearly visible capillary network.Incision yields a characteristic yellowish or gelatinousfluid (Fig. 7.5a) [177, 226].Histologically, an excessive accumulation <strong>of</strong> oedemais a leading microscopic feature. Increased thickness <strong>of</strong><strong>the</strong> walls <strong>of</strong> <strong>the</strong> teleangiectatic blood vessels, <strong>and</strong> thickening<strong>of</strong> <strong>the</strong> epi<strong>the</strong>lial basement membrane complete<strong>the</strong> classical triad <strong>of</strong> morphologic changes. The sulphatedglycosaminoglycans are probably responsible for <strong>the</strong>characteristic blue-coloured abundant amorphous materialin <strong>the</strong> subepi<strong>the</strong>lial stroma in haematoxylin <strong>and</strong>eosin (H&E)-stained slides (Fig. 7.5b) [238]. Fragility<strong>and</strong> alterations in <strong>the</strong> walls <strong>of</strong> blood vessels, such asthin endo<strong>the</strong>lium with many fenestrae <strong>and</strong> vesicles, <strong>and</strong>thickened basement membrane, revealed by electronmicroscopy, are considered important in <strong>the</strong> development<strong>of</strong> RO [320]. Connective tissue proliferation, especiallywith aging <strong>of</strong> <strong>the</strong> lesion, makes <strong>the</strong> lesion irreversibleunless surgical removal is provided. Changes in <strong>the</strong>covering squamous epi<strong>the</strong>lium <strong>of</strong> all three exudative lesionsare expected to be only reactive (squamous cell hyperplasia,basal <strong>and</strong> parabasal cell hyperplasia) <strong>and</strong> mayturn with aging <strong>and</strong> enlargement <strong>of</strong> <strong>the</strong> lesions into atrophicepi<strong>the</strong>lium. Exceptionally, 12 (1.7%) patients withpotentially malignant lesions (atypical hyperplasia, <strong>and</strong>LIN I <strong>and</strong> II) were found in a review <strong>of</strong> two comprehensivestudies. No malignant alteration was reported within<strong>the</strong>se two studies [177, 226].In <strong>the</strong> early stage, only voice rehabilitation <strong>and</strong> avoidance<strong>of</strong> irritating factors should be attempted. However,microlaryngoscopic excision is required in <strong>the</strong> great majority<strong>of</strong> cases. Following surgery, voice <strong>the</strong>rapy is <strong>of</strong>tenindicated [32, 177, 386].7.5.1.2 Vocal Cord Polyp<strong>and</strong> NoduleVocal cord polyp (VCP) <strong>and</strong> vocal cord nodule (VCN)are fairly common benign reactive lesions, causally relatedto phonotrauma <strong>and</strong> vocal abuse [85, 175, 177, 181,194, 308]. The distinction between a VCP <strong>and</strong> VCN is


Larynx <strong>and</strong> Hypopharynx Chapter 7 209abFig. 7.6. Vocal cord polyp. a Huge vocal cord polyp arising from<strong>the</strong> middle third <strong>of</strong> <strong>the</strong> right vocal cord. b Myxomatous fibrous tissuewith fibrin deposition <strong>and</strong> dilated blood vesselsprobably only a matter <strong>of</strong> terminology, since both exhibitsimilar aetiology , pathogenesis , <strong>and</strong> to a certaindegree, similar histologic characteristics.A VCP is a pedunculated or sessile, mono- or multilobulatedlesion, measuring up to 10 mm in diameter,<strong>and</strong> located between <strong>the</strong> anterior <strong>and</strong> middle thirds <strong>of</strong><strong>the</strong> vocal cord [177]. Bilateral polyps are found in only15% <strong>of</strong> cases [21]. They affect primarily adults between20 <strong>and</strong> 50 years <strong>of</strong> age, although <strong>the</strong>y may also occur ino<strong>the</strong>r age groups. Men are affected at least twice as frequentlyas women [177, 181, 194].Vocal chord nodules are smaller, <strong>of</strong>ten bilateral, sessile,fusiform swellings <strong>of</strong> <strong>the</strong> vocal cords, positionedsymmetrically, <strong>and</strong> rarely exceed 2 mm in diameter.They usually occur in children, more frequently in boyswith <strong>the</strong> peak between <strong>the</strong> ages <strong>of</strong> 5 <strong>and</strong> 10 years [363].In adults, <strong>the</strong> highest incidence <strong>of</strong> <strong>the</strong>se lesions is inyoung to middle-aged women. VCNs are considered tobe <strong>the</strong> most common benign lesions <strong>of</strong> <strong>the</strong> vocal cords[177, 386].In addition to phonotrauma <strong>and</strong> vocal abuse, cigarettesmoking , unfavourable occupational exposure , infections<strong>and</strong> endocrine dysfunction , are also consideredto be possible aetiologic factors <strong>of</strong> VCPs [175, 177, 181,194, 308]. VCNs are mainly caused by chronic misuse<strong>and</strong> overuse <strong>of</strong> <strong>the</strong> voice, as well as by emotional disturbancesin children, <strong>and</strong> hormonal disorders <strong>and</strong> smokingin adults [4, 177, 308].Damage <strong>of</strong> <strong>the</strong> subepi<strong>the</strong>lial blood vessels is <strong>the</strong> initialevent in <strong>the</strong> evolution <strong>of</strong> a VCP or VCN. However,<strong>the</strong> morphology <strong>of</strong> <strong>the</strong> two exudative lesions depends on<strong>the</strong> severity <strong>of</strong> <strong>the</strong> initial damage <strong>and</strong> repeated injuries[123, 138, 177, 181, 194, 308].The gross appearance <strong>of</strong> a VCP varies from a glassytranslucent gelatinous formation, to congested <strong>and</strong> purplered in teleangiectatic variants, <strong>and</strong> finally, to whitish,firm <strong>and</strong> opalescent in <strong>the</strong> predominant fibrous forms at<strong>the</strong> end stage <strong>of</strong> <strong>the</strong> lesions (Fig. 7.6a). VCNs start as as<strong>of</strong>t reddish swelling . Gradually, when <strong>the</strong> fibrous tissueproliferates, <strong>the</strong> VCNs become firmer, whitish in colour,<strong>and</strong> conical in shape.Hoarseness is <strong>the</strong> predominant clinical symptom inboth lesions. A great variety <strong>of</strong> voice changes , rangingfrom mild hoarseness to complete aphonia, is found, dependingon <strong>the</strong> location <strong>and</strong> size <strong>of</strong> <strong>the</strong> lesions [21, 94,177, 181, 352].Histologically, different stages <strong>of</strong> VCP developmentare noted. Initially, <strong>the</strong> subepi<strong>the</strong>lial stroma is diffuselyoedematous with dilated vessels. After severe or repeatedinjuries, massive leakage <strong>of</strong> oedema, mainly fibrinas amorphous hyaline pink material, <strong>and</strong> erythrocytes,are <strong>the</strong> predominant features in <strong>the</strong> vicinity <strong>of</strong> <strong>the</strong>angiectatic vessels, which may also be thrombotic (hyalineforms <strong>of</strong> <strong>the</strong> VCP) . Evidently dilated vessels, haemorrhageswith consequent haemosiderosis <strong>and</strong> conspicuousingrowths <strong>of</strong> new blood vessels create <strong>the</strong> angiectaticor vascular stage <strong>of</strong> <strong>the</strong> VCP (Fig. 7.6b) . Finally, <strong>the</strong>lesions may be transformed into a fibrous variant containingan increased amount <strong>of</strong> fibrous tissue <strong>and</strong> bloodvessels. Not infrequently, mixed type VCPs are seen,composed <strong>of</strong> two or more different histologic patterns[21, 123, 177, 194, 308].


210 N. Gale · A. Cardesa · N. Zidar7Fig. 7.7. Intubation granuloma. a Exophytic intubation granulomalocated on <strong>the</strong> posterior third <strong>of</strong> <strong>the</strong> right vocal cord. b Polabypoid granulation tissue is ulcerated with re-epi<strong>the</strong>lialisation at<strong>the</strong> edgeRarely, scattered atypical stromal cells, not associatedwith increased mitotic activity, may be found within <strong>the</strong>core <strong>of</strong> <strong>the</strong> VCP. This finding must not incorrectly leadto a diagnosis <strong>of</strong> malignancy [383].In <strong>the</strong> initial stage, VCNs show diffusely oedematoustissue with distended capillaries <strong>and</strong> venules <strong>and</strong> tinyperivascular haemorrhages surrounded by a minimal ormoderate inflammatory reaction. In time, <strong>the</strong> loose connectivestroma is replaced by a mild to moderate cellularfibrous tissue changing in varying stages <strong>of</strong> evolution.As previously mentioned, <strong>the</strong> covering squamous epi<strong>the</strong>liumin both lesions shows predominantly benignreactive changes. In 4 (0.8%) patients, potentially malignantchanges (atypical hyperplasia) were noted in <strong>the</strong>covering epi<strong>the</strong>lium, but <strong>the</strong>re were no data on malignantalteration [177, 226].The treatment <strong>of</strong> choice for VCPs is microlaryngoscopicsurgical removal. Childhood VCNs may disappearin puberty. Small incipient VCNs in adults may alsovanish spontaneously or after voice rehabilitation . Surgicalintervention is indicated when <strong>the</strong>re is no improvementafter conservative treatment.7.5.2 Contact Ulcer<strong>and</strong> Granuloma,Intubation GranulomaContact ulcer (CU), granuloma (CG) <strong>and</strong> intubationgranuloma (IG) are benign, inflammatory, exophytic orulcerative lesions, usually located in <strong>the</strong> posterior third<strong>of</strong> <strong>the</strong> glottic area. Aetiologically, <strong>the</strong> lesions arise inresponse to various mechanical <strong>and</strong> chemical injuries,such as voice abuse or protracted forceful coughing, acidregurgitation <strong>and</strong> intubation injuries. They display similarsymptomatology <strong>and</strong> clinical appearance, <strong>and</strong> moreor less identical histopathological features <strong>and</strong> prognosis[16, 28, 77, 177, 180, 374, 383, 384].Excessive shouting or coughing cause repeated microtraumas<strong>of</strong> <strong>the</strong> thin mucosa <strong>of</strong> <strong>the</strong> vocal cord processes.They strike each o<strong>the</strong>r in phonatory adduction<strong>of</strong> <strong>the</strong> arytenoids, which leads to <strong>the</strong> development <strong>of</strong> ulcerativeor exophytic lesions <strong>of</strong> one or both vocal cords[384]. Additionally, acid regurgitation due to hiatal herniaor gastritis may cause <strong>the</strong> same type <strong>of</strong> lesions in <strong>the</strong>posterior glottic area.Intubation granuloma is an undesired sequel <strong>of</strong> intubationtube pressure during anaes<strong>the</strong>sia or intensivecare treatment.Intubation granulomas are more common in females,while hyperacidic granulomas <strong>and</strong> CU/CGs are predominantin males [104, 177, 180, 383].Clinically, ulceration or exophytic lesions can befound, mono- or multilobular, frequently bilateral, measuringup to 15 mm in diameter, that range from palegrey to dark red, sometimes with an ulcerated surface(Fig. 7.7a) . Hoarseness , <strong>the</strong> sensation <strong>of</strong> a foreign body ,coughing , a sticking sensation , pain in <strong>the</strong> throat, <strong>and</strong><strong>the</strong> feeling <strong>of</strong> acidity, are <strong>the</strong> prevailing symptoms in allthree types <strong>of</strong> lesions .Histologically, an ulceration <strong>of</strong> <strong>the</strong> posterior mucosa,covered by necrotic tissue <strong>and</strong> fibrin, is initially seen.


Larynx <strong>and</strong> Hypopharynx Chapter 7 211The depth <strong>of</strong> <strong>the</strong> ulcers may vary from superficial todeep lesions extending down to <strong>the</strong> perichondrium <strong>of</strong><strong>the</strong> arytenoid cartilage. The localised necrosis <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lial<strong>and</strong> subepi<strong>the</strong>lial tissue triggers an acute inflammatoryreaction, with proliferation <strong>of</strong> granulation tissueinitially infiltrated by neutrophils <strong>and</strong> later by macrophages,lymphocytes <strong>and</strong> plasma cells (Fig. 7.7b) . Themarginal epi<strong>the</strong>lium starts to proliferate, some regenerativeatypia <strong>of</strong> epi<strong>the</strong>lial cells, such as plump nuclei, <strong>and</strong>increased mitoses may be present [217, 383].An exuberant proliferation <strong>of</strong> granulation tissueforms an exophytic polypoid lesion . New vessels arecharacteristically arranged radially from <strong>the</strong> base to<strong>the</strong> fibrin-covered surface <strong>of</strong> <strong>the</strong> lesion. Approximately1 week after <strong>the</strong> initial injury, connective cells <strong>and</strong> collagenousfibres become more abundant <strong>and</strong> finally <strong>the</strong>predominant elements in <strong>the</strong> granuloma, which in <strong>the</strong>end stage is entirely covered in squamous epi<strong>the</strong>lium .The covering epi<strong>the</strong>lium is usually considerably thickeneddue to hyperplasia <strong>of</strong> <strong>the</strong> prickle cell layer or, rarely,<strong>of</strong> <strong>the</strong> basal <strong>and</strong> parabasal layer [177, 180].The basis <strong>of</strong> <strong>the</strong>rapy in CU/CGs <strong>and</strong> hyperacidicgranulomas is <strong>the</strong> elimination <strong>of</strong> causative factors, voicerest , voice re-education , dietary measures , prohibition<strong>of</strong> smoking <strong>and</strong> alcohol abuse , <strong>and</strong> medical <strong>the</strong>rapy suchas antacids , corticosteroids <strong>and</strong> vitamins [224]. IGs frequentlydo not require treatment due to <strong>the</strong>ir self-limitingnature. In refractory cases, surgical treatment is indicated,ei<strong>the</strong>r microsurgery or CO 2 laser [28].7.5.3 NecrotisingSialometaplasiaNecrotising sialometaplasia (NS) is a rare, benign, selfhealinginflammatory lesion involving <strong>the</strong> minor salivarygl<strong>and</strong>s, primarily <strong>of</strong> <strong>the</strong> hard palate. The lesion isdiscussed in detail in Chap. 5. Here, some specificities<strong>of</strong> <strong>the</strong> extremely rare appearance <strong>of</strong> NS in <strong>the</strong> larynx arepresented [373, 380, 383]. According to previous reports[373, 380], as well as our own experience, NS occurs in<strong>the</strong> larynx secondary to trauma or concomitantly witho<strong>the</strong>r non-neoplastic or neoplastic lesions. The pathogenesisis probably associated with ischaemia. Laryngeal NSappears in <strong>the</strong> supraglottic <strong>and</strong> subglottic regions whereseromucinous gl<strong>and</strong>s are present as a deep ulcerative orsubmucosal nodular lesion. The most prominent histologiccharacteristics that help to distinguish <strong>the</strong> lesion fromvarious forms <strong>of</strong> laryngeal carcinomas are: preservation<strong>of</strong> <strong>the</strong> lobular architecture <strong>of</strong> <strong>the</strong> necrotic gl<strong>and</strong>ular isl<strong>and</strong>s,<strong>the</strong> appearance <strong>of</strong> epi<strong>the</strong>lial-myoepi<strong>the</strong>lial isl<strong>and</strong>swith smooth margins, no cellular atypia or occurrence <strong>of</strong>pathologic mitoses in <strong>the</strong> rest <strong>of</strong> <strong>the</strong> cellular part, <strong>and</strong> <strong>the</strong>retention <strong>of</strong> <strong>the</strong> lumina in preserved ductal formations.The appearance <strong>of</strong> surface pseudoepi<strong>the</strong>liomatous hyperplasiamay cause additional problems in differentialdiagnosis with laryngeal cancers, especially when frozensection analysis is performed. The duration <strong>of</strong> <strong>the</strong> healingprocess is related to <strong>the</strong> size <strong>of</strong> <strong>the</strong> lesion [380].7.5.4 Metaplastic ElasticCartilaginous NodulesMetaplastic elastic cartilaginous nodules (MECN) aresmall (less than 1 cm) fibroelastic lesions occurringmost frequently in <strong>the</strong> posterior <strong>and</strong> mid-portions <strong>of</strong> <strong>the</strong>glottis <strong>and</strong> ventricular b<strong>and</strong>s. Cartilaginous nodules arecomposed <strong>of</strong> a peripheral rim <strong>of</strong> fibroblasts with transitioninto fibroelastic cartilage towards <strong>the</strong> centre [112].Aetiologically, an association with laryngeal trauma hasbeen suggested [277]. The development <strong>of</strong> MECN showsa smooth transition from <strong>the</strong> initial accumulation <strong>of</strong> acidmucopolysaccharides between <strong>the</strong> collagen bundles <strong>and</strong><strong>the</strong>ir separation, to transition <strong>of</strong> fibroblasts to enlarged,rounded cells resembling chondrocytes. Aggregates <strong>of</strong>elastic fibres are present in <strong>the</strong> centre <strong>of</strong> <strong>the</strong> lesions [156].Nodules are usually covered by intact mucosa.Metaplastic elastic cartilaginous nodules are rarelyclinically relevant [277]. We should be aware <strong>of</strong> <strong>the</strong>irpossible existence <strong>and</strong> distinction from chondroma <strong>and</strong>low-grade chondrosarcoma. Chondroma has a characteristiclobular pattern <strong>and</strong> low cellularity, which is not<strong>the</strong> case with MECN. Low-grade chondrosarcomas differmainly from MECN in <strong>the</strong>ir locations, <strong>and</strong> cellular<strong>and</strong> structural atypia [277].7.5.5 AmyloidosisAmyloidosis is a heterogeneous group <strong>of</strong> disorders associatedwith extracellular deposition <strong>of</strong> an abnormalfibrillar protein with pathognomonic tinctorial properties.It may be hereditary or acquired, localised or systemicin distribution. The current classification <strong>of</strong> amyloidosisis based on <strong>the</strong> biochemical composition <strong>of</strong> itspeptide subunits [131].Laryngeal amyloidosis (LA) is rare <strong>and</strong> is mostly a localiseddisease. In <strong>the</strong> majority <strong>of</strong> LA cases, <strong>the</strong> amyloid iscomposed <strong>of</strong> immunoglobulin light chains (AL amyloid).LA may occasionally be part <strong>of</strong> systemic disease or can be associatedwith a tumour, such as neuroendocrine carcinoma<strong>of</strong> <strong>the</strong> larynx or medullary carcinoma <strong>of</strong> <strong>the</strong> thyroid [359].Laryngeal amyloidosis primarily affects patients between40 <strong>and</strong> 60 years <strong>of</strong> age, more frequently males[298]. A few cases have been reported in children [273].All parts <strong>of</strong> <strong>the</strong> larynx can be affected [359], but in somestudies <strong>the</strong> supraglottis was <strong>the</strong> most common site <strong>of</strong> involvement[159]. It can affect <strong>the</strong> larynx multifocally,<strong>and</strong> can also extend to <strong>the</strong> tracheobronchial tree. Themain symptom is hoarseness, in some patients accompaniedby dysphagia, dyspnoea, or haemoptysis [159].


212 N. Gale · A. Cardesa · N. Zidar7Fig. 7.8. Amyloidosis <strong>of</strong> <strong>the</strong> larynx. a Scattered reddish masses <strong>of</strong>amyloid are seen in <strong>the</strong> subepi<strong>the</strong>lial stroma by Congo red stainabing. b The same field, green birefringence <strong>of</strong> <strong>the</strong> amyloid with polarisedlightGrossly, <strong>the</strong> affected area <strong>of</strong> <strong>the</strong> larynx is swollen,sometimes polypoid, covered by an intact mucosa. On acut surface, it is firm, pale, waxy, tan-yellow to red-grey[359]. Microscopically, H&E staining shows <strong>the</strong> deposition<strong>of</strong> an amorphous, eosinophilic, hyaline, extracellularsubstance in <strong>the</strong> subepi<strong>the</strong>lial stroma, blood vesselwalls, <strong>and</strong> along <strong>the</strong> basement membranes <strong>of</strong> seromucinousgl<strong>and</strong>s, with intact covering epi<strong>the</strong>lium. The depositsmay be discrete, or may appear as large roundedmasses <strong>of</strong> variable size. They stain with Congo red <strong>and</strong>display green birefringence with polarised light; thisproperty remains <strong>the</strong> diagnostic gold st<strong>and</strong>ard (Fig. 7.8) .Immunohistochemical analysis must also be performedto determine <strong>the</strong> amyloid type [131].Most patients can be successfully treated by a conservingsurgical excision to preserve laryngeal function for aslong as possible. In some patients, multiple proceduresmay be necessary, <strong>and</strong> recurrences may occur. A fatal outcomehas been described in patients with progressive tracheobronchialinvolvement, but association with systemicamyloidosis is rare [59]. Clinical examination is advisedto exclude <strong>the</strong> possibility <strong>of</strong> a systemic disease.7.5.6 Sinus Histiocytosiswith Massive Lymphadenopathy<strong>and</strong> O<strong>the</strong>r Rare PseudotumoursSinus histiocytosis with massive lymphadenopathy(SHML), or Rosai-Dorfman disease, is an idiopathic,relatively rare benign lesion, based on a nodal <strong>and</strong>/orextranodal histiocytic proliferative disorder that usuallyresolves spontaneously. The most frequent clinicalmanifestation <strong>of</strong> <strong>the</strong> disease is cervical, bilateral<strong>and</strong> painless lymphadenopathy. However, extranodalsites may also be involved <strong>and</strong> <strong>the</strong> head <strong>and</strong> neckregion is one <strong>of</strong> <strong>the</strong> most commonly affected areas[381]. Extranodal disease may be <strong>the</strong> initial or sometimes<strong>the</strong> sole manifestation <strong>of</strong> <strong>the</strong> disease. Foucar<strong>and</strong> co-workers reported that 43% <strong>of</strong> patients had atleast one site <strong>of</strong> extranodal location <strong>of</strong> SHML [118].Within <strong>the</strong> head <strong>and</strong> neck, <strong>the</strong> nasal cavity, paranasalsinuses <strong>and</strong> orbit are commonly involved. Some cases<strong>of</strong> laryngeal lesions have been reported too [5, 11, 52,66, 118, 207]. Typically, <strong>the</strong> SHML begins insidiously<strong>and</strong> progresses to a protracted course <strong>of</strong> <strong>the</strong> activestage, <strong>and</strong> ends with spontaneous remission. SHMLoccasionally appears with subsequent recurrence <strong>and</strong>serious consequences, occasionally even death, if vitalorgans are affected. Exclusive extranodal diseaseis more frequent in elderly persons [51]. LaryngealSHML usually manifests as a circumferential narrowingor polypoid mucosal lesion <strong>of</strong> a tan-white toyellow appearance. Vocal cord involvement results inimpaired mobility [51].Histologically, <strong>the</strong> laryngeal mucosa is almost diffuselyinfiltrated by lymphocytes, plasma cells, neutrophils<strong>and</strong> clusters <strong>of</strong> histiocytes. Histiocytes are <strong>of</strong> varioussizes, with a focally vacuolated pale to pink cytoplasm,but ill-defined borders. Their nuclei are roundor oval, sometimes vesicular, with well-defined centralnucleoli. Lympho- <strong>and</strong> granulophagocytosis is evidentin <strong>the</strong> cytoplasm <strong>of</strong> <strong>the</strong>se histiocytes; <strong>the</strong> phenomenonis termed emperipolesis. Immunohistochemical-


Larynx <strong>and</strong> Hypopharynx Chapter 7 213ly, <strong>the</strong> histiocytes are strongly positive for S-100 protein<strong>and</strong> Leu-M1. No nuclear <strong>and</strong> cytoplasmic atypiais observed.Differential diagnosis includes infectious diseases(rhinoscleroma), Wegener’s granulomatosis, NK/T lymphoma<strong>of</strong> <strong>the</strong> nasal type, eosinophilic granuloma, Hodgkin’slymphoma <strong>and</strong> fibroinflammatory disorders. Rhinoscleromais characterised by a proliferation <strong>of</strong> largemacrophages (Mikulicz cells) in which Klebsiella rhinoscleromatiscan be identified. In this disease <strong>the</strong> phenomenon<strong>of</strong> emperipolesis is not found. Histologically,in Wegener’s granulomatosis <strong>the</strong> S-100 positive histiocytesare lacking, while NK/T lymphoma <strong>of</strong> <strong>the</strong> nasaltype shows an infiltration <strong>of</strong> malignant lymphoidcells. Eosinophilic granuloma is histologically similarto Rosai-Dorfman disease, but differentiation is possiblewith <strong>the</strong> morphologic specificities <strong>of</strong> <strong>the</strong> Langerhanscells, characteristic <strong>of</strong> eosinophilic granuloma: <strong>the</strong>ir nucleishow lobulation, indentation or longitudinal grooving.Finally, fibroinflammatory lesions, such as aggressivefibromatosis, can be easily differentiated due to <strong>the</strong>relatively acellular appearance compared with <strong>the</strong> characteristiccellular infiltration in Rosai-Dorfman disease[381].Some rare laryngeal lesions may clinically <strong>and</strong> pathologicallymimic neoplastic growth <strong>and</strong> could be rangedin a category <strong>of</strong> pseudotumours: hamartoma [282, 314],warty dyskeratoma <strong>of</strong> <strong>the</strong> vocal cord [178], <strong>and</strong> Kimuradisease <strong>of</strong> <strong>the</strong> epiglottis [58].7.5.7 InflammatoryMy<strong>of</strong>ibroblastic TumourICD-O:8825/1Inflammatory my<strong>of</strong>ibroblastic tumour (IMT) is clinicopathologicallya well-defined fibroinflammatory proliferativelesion with unpredictable biological behaviour.Lung, gastrointestinal <strong>and</strong> genitourinary tract systemsare <strong>the</strong> commonest sites for IMT, although <strong>the</strong> lesionhas been reported throughout <strong>the</strong> body [382]. It rarelyaffects <strong>the</strong> head <strong>and</strong> neck region, <strong>and</strong> only a few welldocumented IMTs have been found in <strong>the</strong> larynx <strong>and</strong>pharynx [65, 96, 167, 188, 382].The aetiology <strong>of</strong> <strong>the</strong> lesion is unknown, but differentinfections with an exaggerated response to someunknown microorganism or post-traumatic eventshave been attributed as causal factors [9, 76, 167, 382].Most reported laryngeal IMTs are polypoid or pedunculatedlesions that occur in <strong>the</strong> true vocal cords or in<strong>the</strong> subglottic area. Hoarseness, foreign body sensation,dyspnoea <strong>and</strong> stridor are presenting symptoms.Patients with laryngeal IMTs are mainly adult males[382].Histologically, IMT is composed <strong>of</strong> my<strong>of</strong>ibroblasticspindle cells, admixed with a prominent infiltrateFig. 7.9. Inflammatory my<strong>of</strong>ibroblastic tumour. Lesion is composed<strong>of</strong> uniform spindle cells, intermingled with inflammatorycells<strong>of</strong> lymphocytes, plasma cells <strong>and</strong> neutrophils. The nuclei<strong>of</strong> <strong>the</strong> spindle cells are elongated, slightly polymorphous,containing one or more small nucleoli, <strong>the</strong> cytoplasmis palely eosinophilic (Fig. 7.9) . Occasional regularmitoses are seen. Inflammatory cells are unevenlydistributed within <strong>the</strong> lesion.Three basic histological patterns have been described:1. Myxoid/vascular pattern, resembling inflammatorygranulation tissue;2. Compact spindle cell pattern with fascicular <strong>and</strong>/orstoriform areas with various cellular density;3. Hypocellular pattern, densely collagenised <strong>and</strong> reminiscent<strong>of</strong> a fibrous scar [64].Immunohistochemistry confirms <strong>the</strong> my<strong>of</strong>ibroblasticphenotype <strong>of</strong> <strong>the</strong> spindle cells, which are typically reactiveto vimentin, smooth muscle actin, <strong>and</strong> muscle-specificactin [64, 382]. Additionally, ALK1 <strong>and</strong>/or p80 werereported in a cytoplasmic pattern in 40% <strong>of</strong> cases <strong>of</strong> IMT[56]. Both markers are useful indicators <strong>of</strong> a 2p23 abnormality,suggesting <strong>the</strong> neoplastic nature <strong>of</strong> positive cases<strong>of</strong> IMT. However, it must be interpreted in <strong>the</strong> context <strong>of</strong>histologic <strong>and</strong> o<strong>the</strong>r clinicopathologic data if used as anadjunct to differential diagnosis [56].Radical excision <strong>of</strong> <strong>the</strong> lesions has been reported to becurative in more than 90% <strong>of</strong> extrapulmonary IMTs, includinghead <strong>and</strong> neck lesions [63]. Six out <strong>of</strong> seven patientswith laryngeal IMTs in Wenig’s series were free <strong>of</strong>disease over periods <strong>of</strong> 12 to 36 months after completeexcision. In one patient, laryngectomy was required afterrecurrence <strong>of</strong> <strong>the</strong> disease [382]. A metastatic potentialhas been exceptionally noted in patients with abdominal<strong>and</strong> mediastinal IMTs. However, a fatal outcome<strong>of</strong> a patient with IMT <strong>of</strong> <strong>the</strong> paranasal sinuses has


214 N. Gale · A. Cardesa · N. Zidar7been recently reported [124]. Aggressive behaviour supportsrecent observations that at least a subset <strong>of</strong> IMTsrepresents true neoplasms ra<strong>the</strong>r than reactive my<strong>of</strong>ibroblasticproliferation [124, 165].Differential diagnosis mainly includes spindleshapedlesions, such as spindle cell carcinoma, differenttypes <strong>of</strong> sarcomas <strong>and</strong> lesions composed <strong>of</strong> my<strong>of</strong>ibroblasts<strong>and</strong> fibroblasts. The presence <strong>of</strong> cytokeratinreactivity cannot reliably distinguish between spindlecell carcinoma <strong>and</strong> IMT. In contrast, a lack <strong>of</strong> nuclearatypia <strong>and</strong> considerable mitotic activity favour a diagnosis<strong>of</strong> IMT <strong>and</strong> help to differentiate <strong>the</strong> lesion fromdifferent variants <strong>of</strong> laryngeal malignant tumours.Compared with my<strong>of</strong>ibroblastic lesions, IMTs are generallylarger than nodular fasciitis, tend to occur inyounger age groups <strong>and</strong> are composed <strong>of</strong> longer fascicles<strong>of</strong> spindle cells in an inflammatory backgroundrich in plasma cells [333]. In contrast, nodular fasciitisusually lacks <strong>the</strong> striking inflammatory infiltratecharacteristically present in IMT. The recently describedlaryngeal my<strong>of</strong>ibroblastoma shares many similaritieswith IMT, except for a lack <strong>of</strong> inflammatoryinfiltrate <strong>and</strong> could be considered its pure spindle cellproliferation form [218].7.6 Benign Neoplasms7.6.1 Squamous Cell PapillomaICD-O:8052/0Squamous cell papillomas (SCPs) are <strong>the</strong> most commonbenign epi<strong>the</strong>lial tumours <strong>of</strong> <strong>the</strong> larynx, causally relatedto human papillomavirus (HPV) infection. SCPs arediscussed in detail in Chap. 1.Fig. 7.10. Subglottic haemangioma. A dense proliferation <strong>of</strong> capillariesinfiltrates <strong>the</strong> subepi<strong>the</strong>lial stroma around ductal structurescystic carcinoma <strong>and</strong> mucoepidermoid carcinoma, needto be excluded.7.6.2.2 OncocytomaICD-O:8290/0As has been previously discussed in <strong>the</strong> Sect. 7.2.5, <strong>the</strong>whole spectrum <strong>of</strong> laryngeal oncocytic lesions may beconsidered to be ductal hyperplasia <strong>and</strong> metaplasia witha cystic character ra<strong>the</strong>r than true neoplasms. Solid oncocytoma<strong>of</strong> <strong>the</strong> larynx, similar to those in <strong>the</strong> majorsalivary gl<strong>and</strong>s, probably does not exist in this location.7.6.2 Salivary Gl<strong>and</strong>-Type Tumours7.6.2.1 Pleomorphic AdenomaICD-O:8940/0Pleomorphic adenoma (PA) has rarely been reported in<strong>the</strong> larynx [88, 219, 323]. To date, only about 20 caseshave been described. Men predominate slightly overwomen, <strong>and</strong> <strong>the</strong> patients’ ages range from 15 to 82 years[88]. The supraglottis is by far <strong>the</strong> most common site <strong>of</strong>origin. PA, which may grow up to several centimetresin diameter, occurs as a submucosal mass without ulceration.Histologically, PA shows all <strong>the</strong> characteristics<strong>of</strong> a tumour arising in <strong>the</strong> major salivary gl<strong>and</strong>s (seeChap. 5). The diagnosis <strong>of</strong> laryngeal PA should be consideredvery carefully due to its rarity in this location<strong>and</strong> o<strong>the</strong>r tumours, such as chondrosarcoma, adenoid7.6.3 Haemangioma(Neonatal <strong>and</strong> Adult Types)ICD-O:9120/0Laryngeal haemangiomas (LH) are uncommon lesions<strong>of</strong> vascular origin, defined as benign proliferation <strong>of</strong><strong>the</strong> blood vessels. They are divided into two distinctclinicopathologic entities: <strong>the</strong> neonatal <strong>and</strong> adultforms.Neonatal LH is a rare congenital malformation characteristicallyinvolving <strong>the</strong> subglottic area. Although <strong>the</strong>lesion is present at birth, symptoms become clinicallyevident during <strong>the</strong> first 6 months. A progressive crouplikedisease with inspiratory stridor turns into biphasicstridor as <strong>the</strong> obstruction progresses. Characteristically,<strong>the</strong> symptoms are intermittent, accentuated during crying,when <strong>the</strong> vessels are filled up under increased pressure[195, 196, 290, 312, 328].


Larynx <strong>and</strong> Hypopharynx Chapter 7 215Subglottic LH appears more frequently in girls, witha ratio <strong>of</strong> 2:1 [334]. A co-existence <strong>of</strong> haemangiomas <strong>of</strong><strong>the</strong> skin <strong>and</strong> mucosa <strong>of</strong> <strong>the</strong> oral cavity <strong>and</strong> pharynx, aswell as in o<strong>the</strong>r organs [195, 337], may also be an importantindicator <strong>of</strong> disease.The gross appearance <strong>of</strong> <strong>the</strong> lesion ranges from a flatto polypoid, s<strong>of</strong>t, compressible submucosal mass, pinkreddishto blue in colour. The lesion is usually one-sided,<strong>and</strong> is located in <strong>the</strong> posterolateral subglottic area. However,some haemangiomas are horseshoe-shaped <strong>and</strong> arepresent as a bilateral subglottic reddish swelling [195].The diagnosis is based on characteristic clinical features<strong>and</strong> endoscopic appearance. Biopsy should be avoidedbecause <strong>of</strong> an increased risk <strong>of</strong> excessive bleeding.Laryngeal haemangiomas <strong>of</strong> adults, which are usuallylocalised in <strong>the</strong> glottic <strong>and</strong> supraglottic region, areseen as inconspicuous, submucosal, reddish blue lesions.Common symptoms are hoarseness, dyspnoea <strong>and</strong>/orforeign body sensation [336].Histologically, subglottic LHs are divided into capillary<strong>and</strong> cavernous forms. The majority <strong>of</strong> lesions are<strong>of</strong> <strong>the</strong> capillary type [334], consisting <strong>of</strong> <strong>the</strong> proliferatingcapillaries that infiltrate <strong>the</strong> surrounding submucosalstructures (Fig. 7.10) .Vascular channels may be <strong>of</strong> various sizes, lined withplump endo<strong>the</strong>lial cells in which some regular mitosesmay be present. Vascular tissue is intertwined with fibroustissue <strong>and</strong> infiltrated with a variable amount <strong>of</strong>inflammatory cells. Focally, depositions <strong>of</strong> haemosiderinmay be found in <strong>the</strong> fibrous stroma. It is importantto note that haemangiomas in infants are considerablymore cellular than adult ones [48]. The cavernous form<strong>of</strong> LH is less frequent, composed <strong>of</strong> proliferation <strong>of</strong> largeangiectatic vascular spaces, lined with thin, spindleshapedendo<strong>the</strong>lial cells, <strong>and</strong> filled with erythrocytes.This type <strong>of</strong> lesion is more common in adults. Immunohistochemicalanalysis helps to confirm endo<strong>the</strong>lial cellproliferations in both forms, with positivity for CD31,CD34 <strong>and</strong> factor XIIIa antigens.In differential diagnosis, various lesions <strong>of</strong> vascularorigin, such as <strong>the</strong> vascular type <strong>of</strong> <strong>the</strong> vocal cord polyp(VCP), pyogenic granuloma, intravascular papillaryendo<strong>the</strong>lial hyperplasia <strong>and</strong> even angiosarcoma, mustbe considered. The vascular variant <strong>of</strong> VCP is usuallycharacterised by highly angiectatic vascular spaces surroundedby massive leakage <strong>of</strong> fibrin as amorphous hyalinepink material, a feature not characteristic <strong>of</strong> capillaryhaemangioma. Pyogenic granuloma is diffuselyinfiltrated with neutrophils, which is not <strong>the</strong> case inhaemangiomas, if <strong>the</strong> covering epi<strong>the</strong>lium is intact. Papillaryendo<strong>the</strong>lial hyperplasia represents an organisation<strong>of</strong> thrombosis with papillary proliferation <strong>of</strong> <strong>the</strong> endo<strong>the</strong>lialcells, which is not a striking histological feature<strong>of</strong> haemangioma. The distinction between <strong>the</strong> cellularvariant <strong>of</strong> haemangioma <strong>and</strong> angiosarcoma may sometimesbe problematic. However, anastomoses <strong>of</strong> <strong>the</strong> vascularchannels, lined with considerably pleomorphic endo<strong>the</strong>lialcells with evident pathologic mitoses, certainlyfavour a diagnosis <strong>of</strong> angiosarcoma [48].Subglottic LH <strong>of</strong> infants is generally a self-limited butpotentially fatal lesion, causing progressive airway obstruction.Various treatment modalities have been proposed,including expectant policy, systemic steroids <strong>and</strong>interferon alfa-2a applications, CO 2 laser excision <strong>and</strong>tracheostomy, but no very promising treatments are yetavailable [139, 196, 328].7.6.4 ParagangliomaICD-O:8680/1Paragangliomas are rare benign tumours, originatingfrom <strong>the</strong> anatomically dispersed neuroendocrinesystem (paraganglia), characterised by similar neurosecretorycells derived from <strong>the</strong> neural crest <strong>and</strong>associated with autonomic ganglia. The extra-adrenalportions <strong>of</strong> <strong>the</strong> paraganglia are divided into fourgroups according to <strong>the</strong>ir distribution <strong>and</strong> innervations:branchiomeric, intravagal, aortosympa<strong>the</strong>tic<strong>and</strong> visceral autonomic. The paraganglia <strong>of</strong> <strong>the</strong> larynxare part <strong>of</strong> <strong>the</strong> branchiomeric paraganglionic system,toge<strong>the</strong>r with <strong>the</strong> jugulotympanic, carotid body,subclavian, coronary, aorticopulmonary, pulmonary<strong>and</strong> orbital paraganglia [18].Laryngeal paragangliomas (LP) are extremely rare,almost always benign tumours originating in <strong>the</strong> superior<strong>and</strong> inferior paired paraganglia. The former are localisedin <strong>the</strong> false vocal cords, <strong>the</strong> latter in <strong>the</strong> vicinity<strong>of</strong> <strong>the</strong> cricoid cartilage [18, 255]. Patients are usually <strong>of</strong>middle age with a median age <strong>of</strong> 47 years. Surprisingly,compared with o<strong>the</strong>r neuroendocrine tumours, <strong>the</strong> LPsare three times more common in women. The predominantsite is <strong>the</strong> supraglottic (82%), followed by <strong>the</strong> subglotticin 15% <strong>and</strong> <strong>the</strong> glottic area in 3% <strong>of</strong> cases. Signs<strong>and</strong> symptoms are mainly related to <strong>the</strong> localisation <strong>and</strong>size <strong>of</strong> <strong>the</strong> tumour [45, 289]. LPs are rarely functional,multicentric or associated with o<strong>the</strong>r head <strong>and</strong> neckparagangliomas.Macroscopically, <strong>the</strong> tumour usually presents as arounded submucosal mass with an intact covering mucosa,ranging in size from 0.5 to 6 cm [18]. The tumoursare firm; on <strong>the</strong> cut surface <strong>the</strong>y may be homogenous ornodular, from pink to tan <strong>and</strong> dark red in colour. Prominentvascularity <strong>of</strong> <strong>the</strong> tumour may cause abundantbleeding during biopsy.Histologically, LPs are composed <strong>of</strong> two cell types:chief <strong>and</strong> sustentacular or supporting cells. The chiefcells <strong>of</strong> epi<strong>the</strong>lioid appearance are packed into roundnests showing an organoid pattern, surrounded byhighly vascular fibrous tissue (i.e. Zellballen; Fig. 7.11a) .However, <strong>the</strong> characteristic cell nests may be squeezed<strong>and</strong> not apparent in a small biopsy specimen. The chief


216 N. Gale · A. Cardesa · N. Zidar7Fig. 7.11. Paraganglioma. a Characteristic arrangement into“Zellballen” composed <strong>of</strong> central chief <strong>and</strong> peripheral sustentacabular cells. b Chief cells stain immunohistochemically for synaptophysincells typically have an eosinophilic, finely granular cytoplasm<strong>and</strong> central vesicular nuclei. Cellular pleomorphismmay be present <strong>and</strong> is occasionally prominent,but prognostically unimportant. Rare mitoses can befound, usually less than 2–3 per 10 high power fields.The supporting cells are usually inconspicuous, spindleshaped,<strong>and</strong> most frequently found at <strong>the</strong> edge <strong>of</strong> <strong>the</strong> cellballs [18, 375].Immunohistochemical findings are characteristic<strong>and</strong> decisive for <strong>the</strong> diagnosis. The chief cells are positivefor neuroendocrine markers, such as chromogranin,synaptophysin <strong>and</strong> neuron-specific enolase (Fig. 7.11b) .The paragangliomas usually stain negatively for epi<strong>the</strong>lialmarkers, such as cytokeratin, epi<strong>the</strong>lial membraneantigen, carcinoembryonic antigen, <strong>and</strong> calcitonin.Sustentacular cells are positively stained with S-100protein <strong>and</strong> glial fibrillary acid protein [18, 375].Laryngeal paragangliomas must be primarily differentiatedfrom typical <strong>and</strong> atypical carcinoids. The mostreliable aid is positivity for both epi<strong>the</strong>lial (cytokeratin,epi<strong>the</strong>lial membrane antigen <strong>and</strong> carcinoembryonic antigen)<strong>and</strong> neuroendocrine markers in both types <strong>of</strong> carcinoids[107, 110]. O<strong>the</strong>r, more remote differential diagnosticpossibilities include malignant melanoma, renalcell carcinoma <strong>and</strong> medullary thyroid carcinoma. Melanomacan be confirmed by melan A <strong>and</strong> HMB-45 positivity.Renal cell carcinoma, in contrast to paraganglioma,does not express neuroendocrine markers. Medullarycarcinoma expresses positive staining for calcitonin,amyloid <strong>and</strong> CEA [18, 106, 375].Since paragangliomas are only exceptionally malignant,conservative surgical treatment is suggested [18,20, 110]. There are no histological criteria that could reliablypredict <strong>the</strong> biological behaviour <strong>of</strong> <strong>the</strong> lesion [18].In a recent genetic study, it was postulated that sporadichead <strong>and</strong> neck paragangliomas have deletions at <strong>the</strong>same or closely related loci (11q13 <strong>and</strong> 11q22-23) as <strong>the</strong>irfamily counterparts [33].7.6.5 Granular Cell TumourICD-O:9580/0Granular cell tumour (GCT) is an uncommon benign,slowly growing lesion <strong>and</strong> about half <strong>of</strong> <strong>the</strong> cases occurin <strong>the</strong> head <strong>and</strong> neck region [201, 205, 371]. Varioushistogenetic origins have been attributed to GCT,but recent prevailing opinion supports a relationshipto Schwann cells. The tongue <strong>and</strong> subcutaneous tissue<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck are <strong>the</strong> most common sites <strong>of</strong> <strong>the</strong>tumour, while laryngeal involvement is less frequent[162, 176, 187, 205, 286, 330, 371], <strong>and</strong> comprises about10% <strong>of</strong> all cases [205]. These tumours most commonlyappear in <strong>the</strong> posterior area <strong>of</strong> <strong>the</strong> true vocal cords <strong>and</strong>half <strong>of</strong> <strong>the</strong>m extend into <strong>the</strong> subglottis as a smooth,polypoid <strong>and</strong> sessile lesion [205, 286, 371]. GCT typicallyappears between <strong>the</strong> fourth <strong>and</strong> fifth decades <strong>and</strong><strong>the</strong> average age for laryngeal forms is 36 years [286].The tumour rarely occurs in children [162]. Hoarseness,stridor <strong>and</strong> dysphagia are <strong>the</strong> most common complaints.Histologically, <strong>the</strong> tumour is poorly circumscribed<strong>and</strong> consists <strong>of</strong> clusters <strong>and</strong> sheets <strong>of</strong> rounded <strong>and</strong> polygonalcells with indistinct cellular borders <strong>and</strong> small,bl<strong>and</strong>-looking <strong>and</strong> central nuclei. A mild degree <strong>of</strong> nuclearpleomorphism may be present, but mitotic activityis low. Cytoplasm <strong>of</strong> tumourous cells is abundant,characteristically coarsely granular <strong>and</strong> eosinophilic.


Larynx <strong>and</strong> Hypopharynx Chapter 7 217Cytoplasmic granula are PAS-positive <strong>and</strong> resistant todigestion. Marked desmoplasia is <strong>of</strong>ten present in olderlesions, <strong>the</strong>reby masking <strong>the</strong> presence <strong>of</strong> granularcells. In about 50–60% <strong>of</strong> cases, <strong>the</strong> covering epi<strong>the</strong>liumshows pseudoepi<strong>the</strong>liomatous hyperplasia <strong>of</strong> <strong>the</strong> overlyingsquamous epi<strong>the</strong>lium [190]. This curious histologicalfeature mimicking infiltrative growth <strong>of</strong> isl<strong>and</strong>s <strong>of</strong>squamous epi<strong>the</strong>lium may lead to <strong>the</strong> lesion being mistakenfor a squamous cell carcinoma. However, <strong>the</strong> coexistence<strong>of</strong> GCT <strong>and</strong> true squamous cell laryngeal carcinomahas also been reported [206].Immunohistochemical positivity for S-100 protein,vimentin, CD-68 <strong>and</strong> neuron-specific enolase, <strong>and</strong> negativityfor keratin is in accordance with <strong>the</strong> proposed<strong>the</strong>ory [49, 198, 232]. It is also confirmed by electronmicroscopic examination; <strong>the</strong> cytoplasmic granula werefound to be lysosomal structures that contain infoldings<strong>of</strong> cell membranes similar to those in Schwann cells[245].Differential diagnosis should include benign lesionssuch as rhabdomyoma, paraganglioma or histiocyticproliferations. In contrast to GCT, rhabdomyoma doesnot show infiltrative growth, its cells are larger withwell-defined cellular borders <strong>and</strong> evidence <strong>of</strong> cross-striation.Paraganglioma typically shows an organoid pattern(i.e., Zellballen) <strong>and</strong> positivity for neuroendocrinemarkers. Proliferation <strong>of</strong> histiocytes is usually related toinflammatory reaction. Sheets <strong>of</strong> histiocytes are characteristicallyintermingled with inflammatory cells notcommonly found in GCT. Covering pseudoepi<strong>the</strong>liomatoushyperplasia <strong>of</strong> <strong>the</strong> GCT may lead to incorrect diagnosis<strong>of</strong> squamous cell carcinoma. An identification<strong>of</strong> <strong>the</strong> underlying granular cells may resolve this sometimesdifficult diagnostic problem.Complete surgical excision, with an attempt to preserve<strong>the</strong> normal structures, is <strong>the</strong> treatment <strong>of</strong> choice[371].but <strong>the</strong> tumours may also be asymptomatic. Computerisedtomography is a method <strong>of</strong> choice for radiologicalevaluation [324].Histologically, chondromas show a characteristicwell-defined lobular pattern with benign looking <strong>and</strong>evenly distributed chondrocytes that lack nuclear pleomorphism<strong>and</strong> mitotic activity. The cellularity is judgedto be low, when a given high power (×40) field is unlikelyto contain more than 40 nuclei <strong>of</strong> chondrocytes[83, 209].Pathologic diagnosis <strong>of</strong> laryngeal chondroma, especiallyfrom a small biopsy specimen, should be reportedwith due reservation. A misleading chondroma-likearea may present in a well-differentiated case<strong>of</strong> a chondrosarcoma [26]. It has become apparent thatmany <strong>of</strong> <strong>the</strong> so-called chondromas from <strong>the</strong> past thatrecurred locally were actually misdiagnosed as lowgradechondrosarcomas [83]. It is obvious that <strong>the</strong> distinctionbetween chondroma <strong>and</strong> low-grade chondrosarcomaremains a very difficult task. Increased cellularity,nuclear pleomorphism <strong>and</strong> hyperchromasia, <strong>and</strong><strong>the</strong> appearance <strong>of</strong> clusters <strong>of</strong> malignant-looking chondrocytesin a single lacuna, are <strong>the</strong> most conspicuoushistological features <strong>of</strong> chondrosarcoma. A thoroughexamination <strong>of</strong> <strong>the</strong> entire specimen is suggested, tryingto avoid an incorrect diagnosis <strong>of</strong> a given tumour.Chondromas should also be distinguished from laryngealchondrometaplasia, which appears as small nodules<strong>of</strong> <strong>the</strong> fibroelastic cartilage in <strong>the</strong> submucosal tissue<strong>of</strong> <strong>the</strong> glottic region [112].Local conservative excision is preferred for treatment<strong>of</strong> laryngeal chondromas. Each recurrence <strong>of</strong> <strong>the</strong>lesion should be considered a low-grade chondrosarcoma[358].7.7 Malignant Neoplasms7.6.6 ChondromaICD-O:9220/0Chondromas <strong>of</strong> <strong>the</strong> larynx are exceedingly uncommon,well-circumscribed, small (less than 2 cm) cartilaginoustumours that most commonly originate from <strong>the</strong> posteriorlamina <strong>of</strong> <strong>the</strong> cricoid (70–78%) <strong>and</strong> thyroid cartilage(15–20%) [26, 57, 83, 209, 324, 358], <strong>and</strong> exceptionallyfrom <strong>the</strong> epiglottis [166, 209]. They are morecommon in men than in women, <strong>the</strong> peak incidence rateis in <strong>the</strong> fifth decade [57]. The development <strong>of</strong> chondromasin <strong>the</strong> older population is probably related to <strong>the</strong>alteration <strong>of</strong> <strong>the</strong> ossification process, which starts in <strong>the</strong>cricoid <strong>and</strong> thyroid cartilages in <strong>the</strong> third decade, <strong>and</strong>increases with advanced years [358]. Hoarseness, dyspnoea,dysphagia are <strong>the</strong> usual complaints <strong>of</strong> patients,7.7.1 Potentially Malignant(Precancerous) LesionsPotentially malignant (precancerous) lesions are histologicallydefined as alterations <strong>of</strong> <strong>the</strong> squamous epi<strong>the</strong>liumfrom which invasive SCC develops more <strong>of</strong>ten thanfrom o<strong>the</strong>r epi<strong>the</strong>lial lesions [127, 177, 215]. Differentgrades <strong>of</strong> epi<strong>the</strong>lial lesions that appear during <strong>the</strong> multistepprocess <strong>of</strong> carcinogenesis can easily be identifiedhistologically. They are cumulatively called squamousintraepi<strong>the</strong>lial lesions <strong>of</strong> <strong>the</strong> larynx (SILs) <strong>and</strong> representa wide spectrum <strong>of</strong> histomorphologic changes, rangingfrom benign, reactive lesions, to potentially malignant(risky epi<strong>the</strong>lium) <strong>and</strong> intraepi<strong>the</strong>lial carcinoma. SILsare discussed in detail in Chap. 1.


218 N. Gale · A. Cardesa · N. Zidar77.7.2 Invasive SquamousCell CarcinomaICD-O:8070/3Squamous cell carcinoma (SCC) is by far <strong>the</strong> most commonmalignant tumour <strong>of</strong> <strong>the</strong> larynx <strong>and</strong> hypopharynx,accounting for about 95–96% <strong>of</strong> all malignant tumoursat this location. The majority are conventional-typeSCC.7.7.2.1 EpidemiologySquamous cell carcinoma <strong>of</strong> <strong>the</strong> larynx <strong>and</strong> hypopharynxis <strong>the</strong> second most common respiratory cancer, afterlung cancer [54]. It accounts for 1.6–2% <strong>of</strong> all malignanttumours in men, <strong>and</strong> 0.2–0.4% in women [37,293]. Its incidence is increasing in much <strong>of</strong> <strong>the</strong> world,being slightly higher in urban than in rural areas. It isalso higher among blacks than whites [54, 299].Laryngeal <strong>and</strong> hypopharyngeal SCC occur most frequentlyin <strong>the</strong> sixth <strong>and</strong> seventh decades <strong>of</strong> life. It rarelyoccurs in children <strong>and</strong> adolescents [17, 274]. It is morecommon in men [54, 299], with a male:female ratio <strong>of</strong>about 5:1 worldwide [253]. The male:female ratio is decreasingin some countries, reflecting a greater incidenceamong women. The increasing incidence <strong>of</strong> laryngealcancer in women has been attributed to <strong>the</strong> increasedincidence <strong>of</strong> smoking over <strong>the</strong> last two decades[79].7.7.2.2 AetiologyCigarette smoking <strong>and</strong> alcohol consumption are <strong>the</strong>chief risk factors in laryngeal <strong>and</strong> hypopharyngealcancer, <strong>and</strong> smoking has been shown to have <strong>the</strong> greatesteffect. Epidemiological studies have shown that <strong>the</strong>relative risk <strong>of</strong> laryngeal cancer associated with cigarettesmoking is approximately 10 for all subsites <strong>of</strong> <strong>the</strong>larynx <strong>and</strong> hypopharynx. The role <strong>of</strong> alcohol independent<strong>of</strong> that <strong>of</strong> tobacco is less striking, although plausible[38] <strong>and</strong> is demonstrated in some studies [329].Smoking <strong>and</strong> drinking combined have a multiplicativera<strong>the</strong>r than additive effect [222, 223, 254, 367, 393].Avoidance <strong>of</strong> smoking <strong>and</strong> alcohol consumption couldprevent about 90% <strong>of</strong> <strong>the</strong> current incidence <strong>of</strong> laryngealcancer [101].Some o<strong>the</strong>r factors, such as gastro-oesophageal reflux,diet <strong>and</strong> nutritional factors [38, 98, 120, 200, 276,396] have also been related to an increased risk <strong>of</strong> laryngealcancer development, particularly in patients wholack <strong>the</strong> major risk factors [12, 122].Much attention has recently been paid to <strong>the</strong> possiblerole <strong>of</strong> infection with human papillomavirus (HPV)in <strong>the</strong> pathogenesis <strong>of</strong> laryngeal <strong>and</strong> hypopharyngealcancer, but <strong>the</strong> results are conflicting. HPV, mainly type16, has been found in 3–85% <strong>of</strong> laryngeal cancers [213].Moreover, HPV DNA has been detected in 12–25% <strong>of</strong>individuals with a clinically <strong>and</strong> histologically normallarynx [271, 313]. It appears, <strong>the</strong>refore, that HPV infectionplays little role, if any, in laryngeal carcinogenesis[126, 177, 211, 213].7.7.2.3 Anatomic SitesThe larynx is anatomically divided into three compartments:supraglottic, glottic <strong>and</strong> subglottic. Superiorly<strong>and</strong> posteriorly, it is continuous with <strong>the</strong> hypopharynx.Because <strong>of</strong> this anatomic proximity, <strong>the</strong> tumours <strong>of</strong> <strong>the</strong>larynx <strong>of</strong>ten extend to <strong>the</strong> hypopharynx, as well as viceversa, so that in large tumours, it is impossible to determinewhe<strong>the</strong>r it originates from <strong>the</strong> larynx or hypopharynx.There are geographic differences in <strong>the</strong> topographicdistribution <strong>of</strong> <strong>the</strong> laryngeal SCC [19, 301]. In France,Spain, Italy, Finl<strong>and</strong> <strong>and</strong> <strong>the</strong> Ne<strong>the</strong>rl<strong>and</strong>s, supraglotticSCC predominates, while in <strong>the</strong> USA, Canada, UK <strong>and</strong>Sweden glottic SCC is more common. In Japan, SCC isapproximately equally distributed between <strong>the</strong> two sites.The rarest localisation <strong>of</strong> laryngeal cancer is <strong>the</strong> subglottis(1–5%) [19, 318].Determining <strong>the</strong> primary site <strong>of</strong> origin <strong>of</strong> laryngeal/hypopharyngeal cancer is important as it has a significantimpact on <strong>the</strong> clinical presentation, spread, behaviour,<strong>and</strong> prognosis [117, 300]. This, however is not alwayspossible, especially in large tumours.The most common location <strong>of</strong> supraglottic SCC is <strong>the</strong>epiglottis (45–55% <strong>of</strong> supraglottic cancer; Fig. 7.12a) ,followed by <strong>the</strong> false vocal cords (12–33%) <strong>and</strong> <strong>the</strong> aryepiglotticfolds (8–21%). The remaining cases arise from<strong>the</strong> ventricles <strong>and</strong> <strong>the</strong> arytenoids [19]. Supraglottic SCCtends to spread to oropharynx <strong>and</strong> pyriform sinus, but itrarely invades <strong>the</strong> glottis <strong>and</strong> thyroid cartilage.The most common symptoms in supraglottic cancerare: dysphagia, change in <strong>the</strong> quality <strong>of</strong> <strong>the</strong> voice, a sensation<strong>of</strong> a foreign body in <strong>the</strong> throat, haemoptysis, <strong>and</strong>odynophagia.Lymph node metastases are present in 30–40% <strong>of</strong> patients.The overall 5-year survival rate in supraglotticSCC is 65–75% [19, 301].Glottic SCC arises mostly from <strong>the</strong> anterior half <strong>of</strong> <strong>the</strong>vocal cord or from <strong>the</strong> anterior commissure (Fig. 7.12b) ;a posterior origin is rare. Because <strong>of</strong> poor lymphatic supply,glottic SCC tends to remain localised for a long period.As SCC progresses, it invades <strong>the</strong> vocal muscle resultingin <strong>the</strong> fixed vocal cord, which is an ominous clinicalsign [192]. In <strong>the</strong> late stages <strong>of</strong> <strong>the</strong> disease, it may extendto <strong>the</strong> opposite true vocal cord, to <strong>the</strong> supraglottis<strong>and</strong> subglottis; it may also extend through <strong>the</strong> thyroidcartilage <strong>and</strong> invade <strong>the</strong> s<strong>of</strong>t tissue <strong>of</strong> <strong>the</strong> neck.


Larynx <strong>and</strong> Hypopharynx Chapter 7 219The most common early symptom in glottic cancer ishoarseness. O<strong>the</strong>r symptoms include dysphagia, changein <strong>the</strong> quality <strong>of</strong> <strong>the</strong> voice, a sensation <strong>of</strong> a foreign bodyin <strong>the</strong> throat, haemoptysis <strong>and</strong> odynophagia.The incidence <strong>of</strong> lymph node metastases in <strong>the</strong> earlystages is low (0–11% for T1 <strong>and</strong> T2) <strong>and</strong> increases to 14–40% in <strong>the</strong> advanced stages. The overall 5-year survivalrate is 80–85% [19, 301].If SCC crosses <strong>the</strong> ventricles <strong>and</strong> involves <strong>the</strong> supraglottis<strong>and</strong> subglottis, it is termed transglottic SCC (Fig. 7.12c) .Transglottic carcinoma is rare, accounting for 5% <strong>of</strong> alllaryngeal SCC, <strong>and</strong> is associated with a high incidence <strong>of</strong>lymph node metastases <strong>and</strong> a poor prognosis [235].Subglottic carcinoma is rare. The most common presentingsymptoms are dyspnoea <strong>and</strong> stridor [113], <strong>of</strong>tenrequiring an emergency tracheotomy [350]. The subglotticSCC may spread to <strong>the</strong> thyroid gl<strong>and</strong>, cervicaloesophagus, hypopharynx <strong>and</strong> trachea.About 20–25% <strong>of</strong> patients have cervical lymph nodemetastases at presentation, but about 50% <strong>of</strong> patientshave clinically undetectable metastases in <strong>the</strong> paratracheallymph nodes. It has <strong>the</strong>refore been suggested thatparatracheal <strong>and</strong> superior mediastinal nodes should beremoved in patients with subglottic cancer [149]. A frequentcomplication in <strong>the</strong> course <strong>of</strong> <strong>the</strong> disease is a stomalrecurrence, which is defined as a recurrent SCC in<strong>the</strong> mucocutaneous junction <strong>of</strong> <strong>the</strong> tracheostomy afterlaryngectomy [169, 392, 394]. The overall 5-year survivalrate in subglottic SCC is 40–47% [19, 318].Hypopharyngeal SCC occurs most frequently in <strong>the</strong> pyriformsinus (60–85% <strong>of</strong> hypopharyngeal SCC), <strong>and</strong> rarelyin o<strong>the</strong>r localisations, such as <strong>the</strong> posterior pharyngeal wall(10–20%) <strong>and</strong> postcricoid area (5–15%) [151, 368].Hypopharyngeal cancer frequently extends to <strong>the</strong>larynx.The most frequent symptoms in hypopharyngealSCC are odynophagia, dysphagia <strong>and</strong> a neck mass. O<strong>the</strong>rsymptoms include voice changes, otalgia, <strong>and</strong> constitutionalsymptoms [368].The prognosis <strong>of</strong> hypopharyngeal cancer is poor. Because<strong>of</strong> <strong>the</strong> rich lymphatic supply, unrestricted areafor tumour growth, <strong>and</strong> late symptoms, patients mostlypresent in <strong>the</strong> advanced stages. About 70% <strong>of</strong> patientshave lymph node metastases at presentation. Haematogenousdissemination is ra<strong>the</strong>r frequent (in 20–40% <strong>of</strong>patients) [346]. The overall 5-year survival rate in hypopharyngealSCC is 62.5% [346].ab7.7.2.4 Histological VariantsSeveral variants <strong>of</strong> SCC occur in <strong>the</strong> larynx <strong>and</strong> hypopharynx,including verrucous carcinoma, spindlecell carcinoma, basaloid SCC, papillary SCC, lymphoepi<strong>the</strong>lialcarcinoma, adenoid (acantholytic) SCC <strong>and</strong>adenosquamous carcinoma, which are dealt with sepa-cFig. 7.12. Macroscopic appearance <strong>of</strong> <strong>the</strong> squamous cell carcinoma<strong>of</strong> <strong>the</strong> larynx. a Supraglottic carcinoma: an exophytic tumourat <strong>the</strong> base <strong>of</strong> <strong>the</strong> epiglottis. b Carcinoma <strong>of</strong> <strong>the</strong> vocal cord, endoscopicview. c Transglottic carcinoma crossing <strong>the</strong> ventricles, involvingglottis <strong>and</strong> supraglottis


220 N. Gale · A. Cardesa · N. Zidar7ICD-O:8249/3It is <strong>the</strong> most frequent type <strong>of</strong> NEC in <strong>the</strong> larynx, constituting54% <strong>of</strong> all laryngeal neuroendocrine neoplasms,with approximately 300 cases described in <strong>the</strong> literature[106, 242].Similar to o<strong>the</strong>r types <strong>of</strong> NEC, MD-NEC is morecommon in males, with a wide age range from 20 to83 years. The majority <strong>of</strong> patients are heavy smokers. Itarises mostly in <strong>the</strong> supraglottic region. Hoarseness <strong>and</strong>dysphagia are <strong>the</strong> most common symptoms; 20–30% <strong>of</strong>patients also experience pain [19, 243]. MD-NEC is rarelyassociated with carcinoid syndrome [388]. Some paratelyin Chap. 1. Their recognition is important becausemost <strong>of</strong> <strong>the</strong>m are true clinicopathologic entities, with animportant prognostic implication: basaloid SCC, adenosquamouscarcinoma <strong>and</strong> lymphoepi<strong>the</strong>lial carcinomaare more aggressive than conventional SCC, while verrucousSCC <strong>and</strong> arguably, papillary SCC, have a betterprognosis than conventional SCC.7.7.2.5 TNM GradingFor <strong>the</strong> staging <strong>of</strong> laryngeal cancer, <strong>the</strong> TNM system(T – tumour, N – node, M – metastasis), establishedby <strong>the</strong> International Union Against Cancer (UICC) iswidely used [340]. Stage remains <strong>the</strong> most significantpredictor <strong>of</strong> survival. The size <strong>of</strong> <strong>the</strong> tumour <strong>and</strong> <strong>the</strong>presence <strong>of</strong> regional <strong>and</strong> distant metastases are independentpredictors <strong>of</strong> survival.An important parameter ignored in <strong>the</strong> TNM systemis extracapsular spread. Several studies have shown that<strong>the</strong> presence <strong>of</strong> extracapsular spread in <strong>the</strong> lymph nodesis strongly associated with both regional recurrence <strong>and</strong>distant metastases, resulting in decreased survival [71,116, 157, 344]. Some studies, on <strong>the</strong> contrary, have notconfirmed <strong>the</strong> independent prognostic significance <strong>of</strong>extracapsular spread [225, 292].7.7.3 Neuroendocrine CarcinomaNeuroendocrine carcinomas (NEC) <strong>of</strong> <strong>the</strong> larynx areuncommon, accounting for less than 1% <strong>of</strong> laryngealtumours. NEC are divided into well-differentiated NEC(WD-NEC; carcinoid), moderately differentiated NEC(MD-NEC; atypical carcinoid), <strong>and</strong> poorly differentiatedNEC (PD-NEC; small cell carcinoma). MD-NEC is<strong>the</strong> most common type <strong>of</strong> NEC in <strong>the</strong> larynx, followedby PD-NEC <strong>and</strong> WD-NEC [19, 92, 106].The putative cells <strong>of</strong> origin are <strong>the</strong> Kultschitzky-likeargyrophilic cells, which have been described in <strong>the</strong> humanlaryngeal mucosa, <strong>and</strong> are similar or identical to<strong>the</strong> Kultschitzky cells in <strong>the</strong> bronchial mucosa [285,288]. O<strong>the</strong>r possible cells <strong>of</strong> origin are <strong>the</strong> pluripotentstem cells <strong>of</strong> <strong>the</strong> surface or gl<strong>and</strong>ular epi<strong>the</strong>lium.7.7.3.1 Well-DifferentiatedNeuroendocrine Carcinoma(Carcinoid)ICD-O:8240/3Well-differentiated neuroendocrine carcinoma (WD-NEC) is <strong>the</strong> least common type <strong>of</strong> laryngeal NEC. Ina critical review <strong>of</strong> <strong>the</strong> world literature, El-Naggar <strong>and</strong>Batsakis found only 12 well-documented cases <strong>of</strong> laryngealWD-NEC [91]. Since <strong>the</strong>n, a few more cases havebeen described in <strong>the</strong> English-language literature [67,191, 242, 326, 341, 391].Well-differentiated NEC occurs predominantlyin males, <strong>the</strong> average age is 58 years, <strong>and</strong> <strong>the</strong> majority(83%) are located in <strong>the</strong> supraglottis [92]. They presentclinically with dyspnoea, hoarseness <strong>and</strong>/or sore throat.Grossly, laryngeal WD-NEC is typically a submucosalnodule or a polypoid lesion measuring up to2 cm in diameter. Microscopically, like WD-NEC elsewherein <strong>the</strong> body, <strong>the</strong>y are composed <strong>of</strong> small uniformcells growing in isl<strong>and</strong>s, ribbons <strong>and</strong> cords, occasionallyforming gl<strong>and</strong>-like structures. Mucin may occasionallybe present. The nuclei are round, with finelydispersed chromatin <strong>and</strong> inconspicuous nucleoli; <strong>the</strong>cytoplasm is scant, clear or eosinophilic. Mitoses aresparse or absent, <strong>and</strong> <strong>the</strong>re is no necrosis or cellularpleomorphism.Immunohistochemically, laryngeal WD-NEC expressesmarkers <strong>of</strong> neuroendocrine differentiation (suchas chromogranin, synaptophysin, neuron-specific enolase,Leu-7), <strong>and</strong> markers <strong>of</strong> epi<strong>the</strong>lial differentiation(such as cytokeratins <strong>and</strong> epi<strong>the</strong>lial membrane antigen)[106]. Electron microscopy reveals dense-core neurosecretorygranules [91, 106].Differential diagnosis includes moderately differentiatedNEC, paraganglioma, <strong>and</strong> adenocarcinoma, <strong>and</strong> isdiscussed in <strong>the</strong> next section.The treatment <strong>of</strong> choice is complete but conservativesurgical excision. <strong>Neck</strong> dissection is not indicated. Radio<strong>the</strong>rapy<strong>and</strong> chemo<strong>the</strong>rapy have not proven effective[106, 248].The prognosis is favourable, although metastases to<strong>the</strong> lymph node, liver, bones <strong>and</strong> skin have been reportedin one-third <strong>of</strong> patients. Only one patient has died <strong>of</strong><strong>the</strong> disease [91]. These data suggest <strong>the</strong> more aggressivebehaviour <strong>of</strong> laryngeal WD-NEC compared with bronchialWD-NEC, but <strong>the</strong> number <strong>of</strong> patients is too smallto draw any conclusions [19].7.7.3.2 Moderately DifferentiatedNeuroendocrine Carcinoma(Atypical Carcinoid)


Larynx <strong>and</strong> Hypopharynx Chapter 7 221Fig. 7.13. Moderately differentiated neuroendocrine carcinoma<strong>of</strong> <strong>the</strong> larynx. a Isl<strong>and</strong>s <strong>of</strong> closely packed small cells with hyperabchromatic nuclei beneath <strong>the</strong> surface squamous cell epi<strong>the</strong>lium. bImmunohistochemical expression <strong>of</strong> CD56 in tumour cellstients with MD-NEC have an elevated level <strong>of</strong> <strong>the</strong> serumcalcitonin [23, 338].Grossly, it presents as a submucosal nodule or as apolypoid lesion measuring up to 4 cm in diameter (average1.6 cm), with or without surface ulceration.Microscopically, <strong>the</strong> tumour grows in rounded nests,trabeculae, cords, ribbons <strong>and</strong> gl<strong>and</strong>ular structures; <strong>the</strong>tumour cells are round, with round nuclei <strong>and</strong> a moderateamount <strong>of</strong> cytoplasm, which is slightly eosinophilicor occasionally oncocytic. Mucin production may bepresent [234].In contrast to WD-NEC, cellular pleomorphism, increasedmitotic activity <strong>and</strong> necroses are frequentlypresent in MD-NEC. Vascular <strong>and</strong> perineural invasionmay be present.Immunohistochemically, MD-NEC usually expressessynaptophysin, cytokeratin, <strong>and</strong> chromogranin; <strong>the</strong>ymay also express CD56, calcitonin <strong>and</strong> carcinoembryonicantigens, but rarely serotonin (Figs. 7.13) [97, 234,242, 388].Differential diagnosis includes paraganglioma, adenocarcinoma,o<strong>the</strong>r neuroendocrine carcinomas <strong>and</strong>medullary carcinoma <strong>of</strong> <strong>the</strong> thyroid gl<strong>and</strong>.The differentiation between paraganglioma <strong>and</strong> MD-NEC is important because <strong>the</strong> former usually behaves asa benign tumour, while <strong>the</strong> latter behaves as an aggressivetumour. The correct diagnosis is usually possible with <strong>the</strong>use <strong>of</strong> immunohistochemistry: MD-NEC expresses cytokeratin<strong>and</strong> carcinoembryonic antigen (CEA), whileparaganglioma does not. Both tumours express markers<strong>of</strong> neuroendocrine differentiation [18, 242]. Adenocarcinomacan be distinguished from carcinoid by <strong>the</strong> absence<strong>of</strong> neuroendocrine markers. The presence <strong>of</strong> cellular pleomorphism,increased mitotic activity <strong>and</strong> necroses helpsto distinguish MD-NEC from WD-NEC.Differentiation from thyroid medullary carcinomamay be difficult, especially when dealing with cervicalmetastases, as tumour cells in both medullary carcinoma<strong>and</strong> MD-NEC express calcitonin by immunohistochemistry.The most important distinguishing featureis <strong>the</strong> different locations <strong>of</strong> <strong>the</strong> primary tumours. Additionaluseful information may be obtained by measuring<strong>the</strong> serum level <strong>of</strong> CEA, which is elevated in metastaticmedullary carcinoma <strong>of</strong> <strong>the</strong> thyroid, <strong>and</strong> normalin MD-NEC [19]. The elevated serum level <strong>of</strong> calcitoninshould not be considered as a reliable feature <strong>of</strong> medullarycarcinoma, as it has been reported in patients withMD-NEC [92, 338].Moderately differentiated NEC is an aggressive, potentiallylethal tumour. Lymph node metastases havebeen reported in 43% <strong>of</strong> patients, cutaneous metastasesin 22% <strong>and</strong> distant metastases in 44% <strong>of</strong> patients, mostlyto <strong>the</strong> lungs, liver <strong>and</strong> bones [97, 234, 388].Surgery is <strong>the</strong> treatment <strong>of</strong> choice. <strong>Neck</strong> dissectionis also advised because <strong>of</strong> <strong>the</strong> high incidence <strong>of</strong> cervicallymph node metastases. Radiation <strong>and</strong> chemo<strong>the</strong>rapyhave not been effective [248]. The 5- <strong>and</strong> 10-year survivalrates are 48 <strong>and</strong> 30% respectively [388].7.7.3.3 Poorly DifferentiatedNeuroendocrine Carcinoma(Small Cell Carcinoma)ICD-O:8041/3Poorly differentiated neuroendocrine carcinoma is <strong>the</strong>least differentiated <strong>and</strong> <strong>the</strong> most aggressive type <strong>of</strong> NEC.It is rare, accounting for less than 0.5% <strong>of</strong> all laryngealcarcinomas. Approximately 160 cases have been describedin <strong>the</strong> literature [106, 244].


222 N. Gale · A. Cardesa · N. Zidar7Poorly differentiated NEC arises most <strong>of</strong>ten in <strong>the</strong>supraglottis, but it also occurs in o<strong>the</strong>r parts <strong>of</strong> <strong>the</strong> larynx.It affects men more frequently than women, mostlybetween 50 <strong>and</strong> 70 years <strong>of</strong> age; most patients are heavysmokers. The most common presenting symptoms arehoarseness <strong>and</strong> dysphagia, frequently associated withpainless enlarged cervical lymph nodes due to metastases.It may be associated with a paraneoplastic syndrome[108, 244].Grossly, PD-NECs are submucosal nodular or polypoidmasses, frequently ulcerated <strong>and</strong> cannot be distinguishedfrom o<strong>the</strong>r laryngeal carcinomas.Microscopically, laryngeal PD-NECs are identicalto <strong>the</strong>ir pulmonary counterparts [243]. They are composed<strong>of</strong> closely packed small cells with hyperchromaticround, oval or spindle nuclei <strong>and</strong> very scant cytoplasm.Necroses, mitoses, as well as vascular <strong>and</strong> perineural invasionare frequently present. PD-NEC can also be composed<strong>of</strong> slightly larger cells with more cytoplasm. Themucosa is <strong>of</strong>ten ulcerated, but <strong>the</strong>re is no carcinoma insitu or significant atypia.Immunohistochemically, <strong>the</strong> tumour cells variablyexpress cytokeratins <strong>and</strong> neuroendocrine markers, suchas synaptophysin, neuron-specific enolase, chromogranin,S-100 protein <strong>and</strong> CD56.By electron microscopy, sparse neurosecretory granulesare occasionally found, but <strong>the</strong>y may be absent.In <strong>the</strong> differential diagnosis, <strong>the</strong> possibility <strong>of</strong> a metastasisfrom <strong>the</strong> lung must be excluded. PD-NEC mustalso not be confused with <strong>the</strong> basaloid squamous carcinoma,malignant lymphoma, <strong>and</strong> malignant melanoma.Basaloid squamous carcinoma is composed <strong>of</strong>larger cells, contains areas <strong>of</strong> squamous differentiation,tends to stain for high molecular-weight cytokeratins,<strong>and</strong> is frequently associated with atypia <strong>of</strong> <strong>the</strong>overlying squamous epi<strong>the</strong>lium. Malignant lymphomascharacteristically express leukocyte common antigen<strong>and</strong> B- or T-cell markers, which are absent in PD-NEC. Malignant melanoma occasionally consists <strong>of</strong>small undifferentiated cells, thus resembling PD-NEC,but, in contrast to PD-NEC, it typically expresses S-100protein, melan A <strong>and</strong>/or HMB45.The clinical course is aggressive, characterised byearly metastases to <strong>the</strong> regional lymph nodes <strong>and</strong> distantsites, especially to <strong>the</strong> lungs, bones <strong>and</strong> liver. In contrastto lung PD-NEC, laryngeal PD-NEC does not frequentlymetastasise to <strong>the</strong> brain.Radiation with chemo<strong>the</strong>rapy is <strong>the</strong> treatment <strong>of</strong>choice. Surgical <strong>the</strong>rapy is not indicated because mostpatients have disseminated disease at presentation. Theprognosis is poor, <strong>and</strong> <strong>the</strong> 2- <strong>and</strong> 5-year survival ratesare 16 <strong>and</strong> 5% respectively [114, 135].7.7.4 AdenocarcinomaIn spite <strong>of</strong> ra<strong>the</strong>r prominent salivary gl<strong>and</strong> tissue in <strong>the</strong>supraglottic <strong>and</strong> subglottic larynx, laryngeal adenocarcinomais rare, accounting for 1% <strong>of</strong> all laryngealneoplasms [3, 25, 347]. The majority <strong>of</strong> laryngeal adenocarcinomasare <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong> type. The mostcommon types are adenoid cystic carcinoma <strong>and</strong> mucoepidermoidcarcinoma. Rare examples <strong>of</strong> o<strong>the</strong>r types <strong>of</strong>adenocarcinoma have been also described in <strong>the</strong> larynx,such as acinic cell carcinoma [306], clear cell carcinoma[262], malignant myoepi<strong>the</strong>lioma [168], epi<strong>the</strong>lial-myoepi<strong>the</strong>lialcarcinoma [240], salivary duct carcinoma[109], etc.The aetiology is unknown, although exposure to asbestosor lead, alcohol abuse, viral infections, ionisingradiation <strong>and</strong> genetic risk factors have been implicatedas possible aetiologic factors [221].7.7.4.1 Adenoid Cystic CarcinomaICD-O:8200/3In contrast to o<strong>the</strong>r laryngeal carcinomas, adenoidcystic carcinoma (ACC) occurs at a younger age, withno gender predominance, <strong>and</strong> is more common in <strong>the</strong>subglottis [348]. Symptoms are similar to those <strong>of</strong> o<strong>the</strong>rtumours in <strong>the</strong> same localisation. In addition, pain isfrequently present, probably because <strong>of</strong> <strong>the</strong> tendency <strong>of</strong>ACC towards perineural invasion.Macroscopically, <strong>the</strong> tumour usually grows as a submucosalmass, covered by normal mucosa.The microscopic features <strong>of</strong> laryngeal ACC are <strong>the</strong>same as in o<strong>the</strong>r locations.Laryngeal ACC is characterised by a slowly progressivecourse, with a high incidence <strong>of</strong> local recurrence,long survival <strong>and</strong> a low cure rate. ACC has a tendencytowards haematogenic spread, mostly to <strong>the</strong> lungs<strong>and</strong> less frequently to <strong>the</strong> bones, liver <strong>and</strong> o<strong>the</strong>r organs[86, 348]. It does not usually metastasise to <strong>the</strong> regionallymph nodes.The treatment <strong>of</strong> choice is complete surgical excision.The 5-year survival rate is 30% [221, 347].7.7.4.2 Mucoepidermoid CarcinomaICD-O:8430/3Mucoepidermoid carcinoma (MEC) occurs at all ages,even in childhood, but it usually presents in <strong>the</strong> 6th <strong>and</strong>7th decades, predominantly in males. The majority occursin <strong>the</strong> supraglottis, but it has also been described in<strong>the</strong> glottis <strong>and</strong> subglottis, as well as in <strong>the</strong> hypopharynx[221, 335]. The clinical picture correlates with <strong>the</strong> localisation<strong>and</strong> size <strong>of</strong> <strong>the</strong> tumour.


Larynx <strong>and</strong> Hypopharynx Chapter 7 223aFig. 7.14. Chondrosarcoma <strong>of</strong> <strong>the</strong> cricoid cartilage <strong>of</strong> <strong>the</strong> larynx.a Typical macroscopic appearance <strong>of</strong> <strong>the</strong> tumour located in <strong>the</strong>cricoid cartilage: cut section reveals a glistening, lobulated, glassybtumour. b Microscopically, <strong>the</strong>re is slightly increased cellularity,binucleation in <strong>the</strong> lacunar spaces, mild nuclear pleomorphism<strong>and</strong> hyperchromasiaMacroscopically, <strong>the</strong>y mostly present as submucosalmasses [335].Microscopically, <strong>the</strong>y are similar to MEC in o<strong>the</strong>rsites, <strong>and</strong> are classified as low-, intermediate-, <strong>and</strong> highgradeMEC (see Chap. 5).The behaviour is unpredictable, <strong>and</strong> is related to <strong>the</strong>grade <strong>and</strong> stage <strong>of</strong> <strong>the</strong> disease. The best treatment iscomplete surgical excision. Radio<strong>the</strong>rapy has been reportedto be successful in a limited number <strong>of</strong> patients[335]. <strong>Neck</strong> dissection may be necessary, as 50% <strong>of</strong> patientswith MEC have metastases in <strong>the</strong> regional lymphnodes. The 5-year survival is 90–100% for low-gradeMEC, <strong>and</strong> 50% for high-grade MEC [115].7.7.5 SarcomasSarcomas <strong>of</strong> <strong>the</strong> larynx are uncommon, accounting for1–2% <strong>of</strong> all laryngeal neoplasms. Among <strong>the</strong>m, chondrosarcomais <strong>the</strong> most frequent type, comprising 75%<strong>of</strong> all laryngeal sarcomas [209].7.7.5.1 ChondrosarcomaICD-O:9220/3Chondrosarcoma (CS) is <strong>the</strong> most common non-epi<strong>the</strong>lialneoplasm in <strong>the</strong> larynx. It appears that laryngeal CSbehaves less aggressively than its counterpart in <strong>the</strong> rest<strong>of</strong> <strong>the</strong> body. The majority <strong>of</strong> laryngeal CS are low gradeCS [209, 360].Laryngeal CS affects men more frequently than women,mostly in <strong>the</strong> 7th decade [209]. It usually presentswith hoarseness; o<strong>the</strong>r symptoms include dyspnoea,dysphonia, a cough, a neck mass, airway obstruction<strong>and</strong> pain [209, 360]. The symptoms are frequently presentfor a long time before <strong>the</strong> diagnosis is established.Chondrosarcoma arises predominantly in <strong>the</strong> cricoidcartilage, especially at <strong>the</strong> inner posterior plate; it canalso arise in <strong>the</strong> thyroid <strong>and</strong> arytenoid cartilages. It veryrarely arises in <strong>the</strong> epiglottis [209, 360].The aetiology is unknown, although disordered ossification<strong>of</strong> <strong>the</strong> laryngeal cartilages <strong>and</strong> ischaemic changesin a chondroma have been suggested as possible predisposingrisk factors [360]. O<strong>the</strong>r possible risk factorsinclude previous radiation exposure [134] <strong>and</strong> Teflon injection[147].Grossly, CS is characteristically a lobulated, submucosalmass covered by normal mucosa; on its cut surfaceit is glassy, firm white or grey (Fig. 7.14a) . Radiographicfindings are characteristic showing coarse or stippledcalcifications [360, 377]. Microscopically, laryngealCS is indistinguishable from CS <strong>of</strong> bone origin elsewherein <strong>the</strong> body, <strong>and</strong> is graded according to <strong>the</strong> histologiccriteria proposed by Evans <strong>and</strong> co-workers [99]for CS <strong>of</strong> <strong>the</strong> bones. Low-grade CS (grade I) has slightlyincreased cellularity, binucleation in <strong>the</strong> lacunar spaces,slight nuclear pleomorphism, <strong>and</strong> hyperchromasia(Fig. 7.14b) . High-grade CS (grade III) has remarkablecellularity, multinucleation in <strong>the</strong> lacunar spaces, nuclearpleomorphism, nuclear hyperchromasia, necrosis<strong>and</strong> mitotic activity, whereas <strong>the</strong> intermediate grade CS(grade II) has medium cellularity <strong>and</strong> less nuclear pleomorphism[128].The vast majority <strong>of</strong> laryngeal CS are <strong>of</strong> low- or intermediategrade. High-grade CS are considered to be rare;in a large series <strong>of</strong> 111 laryngeal CS, only 6 (6%) were <strong>of</strong>a high grade [360]. Dedifferentiated (mesenchymal) CS


224 N. Gale · A. Cardesa · N. Zidar7characterised by <strong>the</strong> presence <strong>of</strong> both well-differentiatedCS <strong>and</strong> a high-grade non-cartilaginous sarcoma is evenrarer [46, 128, 259, 360].Immunohistochemically, CS expresses S-100 protein<strong>and</strong> vimentin [46].Differential diagnosis includes chondroma <strong>and</strong> chondrometaplasia.Differentiation between low-grade CS<strong>and</strong> chondroma can be extremely difficult, <strong>and</strong> is onlypossible with adequate tumour sampling. Chondromasare considered to be exceedingly rare in <strong>the</strong> larynx; <strong>the</strong>yare smaller than CS <strong>and</strong> less cellular, with less pleomorphism,lacking mitoses <strong>and</strong> necrosis.The treatment <strong>of</strong> choice is conservative surgery [46,182, 209]. Total laryngectomy should be avoided as longas possible, even in recurrent CS. Radiation <strong>the</strong>rapy isgenerally regarded as ineffective though a few cases witha favourable response to radiation have been reported[141].The prognosis is favourable. CS is characterised bya slowly progressive growth, with frequent recurrences(18–40%) that are related to incomplete surgical excision<strong>and</strong>/or higher tumour grade [360]. Metastases fromlaryngeal CS are unusual <strong>and</strong> are reported in approximately10% <strong>of</strong> patients, most commonly to <strong>the</strong> lungs<strong>and</strong> lymph nodes [259, 360]. The 5- <strong>and</strong> 10-year survivalrates are 90 <strong>and</strong> 80.9% respectively.7.7.5.2 O<strong>the</strong>r SarcomasRare examples <strong>of</strong> o<strong>the</strong>r sarcomas have been described in<strong>the</strong> larynx <strong>and</strong> hypopharynx, such as liposarcoma [136],osteosarcoma [220, 256, 332], angiosarcoma [214], synovialsarcoma [34], malignant fibrous histiocytoma [143],Kaposi’s sarcoma [246, 325], leiomyosarcoma [202, 297],etc.The aetiology <strong>of</strong> laryngeal sarcomas is unknown, althoughexposure to radiation [284] has been implicatedas a possible aetiologic factor for osteosarcoma [332]<strong>and</strong> malignant fibrous histiocytoma [143], <strong>and</strong> infectionwith HIV for Kaposi’s sarcoma [246, 325].7.7.6 O<strong>the</strong>r Malignant NeoplasmsThe larynx may be rarely involved in disseminatedsystemic lymphoma or leukaemia [163]. It can also be<strong>the</strong> primary site <strong>of</strong> a haematopoietic or lymphoid neoplasm.Extramedullary plasmacytoma seems to be <strong>the</strong>most common primary lymphoid neoplasm <strong>of</strong> <strong>the</strong> larynx.Various types <strong>of</strong> non-Hodgkin’s lymphoma <strong>of</strong> B-cell type <strong>and</strong> T-cell type have also been reported in <strong>the</strong>larynx, as well as rare cases <strong>of</strong> granulocytic sarcoma <strong>and</strong>mast cell sarcoma [163].7.7.6.1 Malignant LymphomaPrimary non-Hodgkin’s lymphoma (NHL) <strong>of</strong> <strong>the</strong> larynxis rare, accounting for less than 1% <strong>of</strong> all laryngeal neoplasms[8] <strong>and</strong> approximately 1% <strong>of</strong> all primary extranodallymphomas [389]. By definition, <strong>the</strong> bulk <strong>of</strong> <strong>the</strong>disease should occur in <strong>the</strong> larynx [389]. About 65 caseshave been reported in <strong>the</strong> English-language literature [8,55, 70, 249].The majority <strong>of</strong> laryngeal NHL are <strong>of</strong> B-cell type, especiallydiffuse large B-cell lymphoma, <strong>and</strong> extranodalmarginal lymphoma <strong>of</strong> <strong>the</strong> MALT type. Rare cases <strong>of</strong> T-cell NHL have been reported, such as NK/T cell lymphoma<strong>of</strong> <strong>the</strong> nasal type, <strong>and</strong> peripheral T-cell lymphoma[389]. All regions <strong>of</strong> <strong>the</strong> larynx may be involved, with<strong>the</strong> exception <strong>of</strong> <strong>the</strong> extranodal marginal lymphoma <strong>of</strong><strong>the</strong> MALT type, which has been described in <strong>the</strong> supraglottisonly [70, 199], presumably because mucosa-associatedlymphoid tissue has been found mostly in <strong>the</strong> supraglotticregion [199].They present mostly at stage I or II <strong>and</strong> are limitedto <strong>the</strong> larynx, with or without regional lymph node involvement.The most common symptoms are hoarseness,foreign body sensation, <strong>and</strong> airway obstruction.The prognosis is favourable. Laryngeal NHL shouldbe treated according to <strong>the</strong> histologic type <strong>of</strong> NHL. Itusually responds well to radiation <strong>the</strong>rapy. Systemicchemo<strong>the</strong>rapy is indicated for recurrent or disseminateddisease [8].7.7.6.2 Extraosseous(Extramedullary) PlasmacytomaICD-O:9734/3Extraosseous plasmacytoma is a clonal proliferation <strong>of</strong>plasma cells forming a tumour at an extraosseous <strong>and</strong>extramedullary site. By definition, <strong>the</strong>re is no evidence<strong>of</strong> plasma cell myeloma on bone marrow examinationor by radiography [142]. The malignant plasma cellsexpress monotypic cytoplasmic immunoglobulins, <strong>and</strong>plasma cell-associated antigens, with an absence <strong>of</strong> immatureB-cell antigens [142, 389].The majority <strong>of</strong> extraosseous plasmacytomas occurin <strong>the</strong> upper respiratory tract; among <strong>the</strong>m, <strong>the</strong> larynxis involved in only 6–18%. An incidence <strong>of</strong> 1–5 plasmacytomasin 1,000 laryngeal tumours has been reported[269].Laryngeal plasmacytoma is more frequent in males,with a mean age <strong>of</strong> 60 years. The epiglottis is <strong>the</strong> mostcommon site <strong>of</strong> involvement, followed by <strong>the</strong> vocalcords, false cords, ventricles <strong>and</strong> <strong>the</strong> subglottis [183, 269,270, 303, 378]. It generally presents as a solitary, submucosallesion, or as a polypoid lesion. It may occasionallyinvolve multiple sites in <strong>the</strong> larynx [269, 303].


Larynx <strong>and</strong> Hypopharynx Chapter 7 225Histologically, <strong>the</strong> tumour consists <strong>of</strong> sheets <strong>of</strong> plasmacells that vary in differentiation from well- to poorlydifferentiated. They may contain Russell bodies orgrape-like inclusions <strong>of</strong> retained immunoglobulin (Mottcells), which are also found in reactive plasma cells <strong>and</strong>do not help in establishing <strong>the</strong> diagnosis <strong>of</strong> plasmacytoma[142].Well-differentiated plasmacytoma cannot be distinguishedmorphologically from reactive (polyclonal) proliferation<strong>of</strong> plasma cells. Therefore, <strong>the</strong> monoclonality<strong>of</strong> plasma cells must be proven, which is best achieved bydemonstrating <strong>the</strong> cytoplasmic immunoglobulin heavy<strong>and</strong>/or light chain restriction. Besides immunohistochemistry,non-isotopic paraffin section in situ hybridisationis useful in <strong>the</strong> assessment <strong>of</strong> clonality for kappaor lambda light chain mRNA [389].Poorly differentiated plasmacytoma must be differentiatedfrom o<strong>the</strong>r lymphoid neoplasms <strong>and</strong> from o<strong>the</strong>rmalignant tumours, such as malignant melanoma <strong>and</strong>carcinoma. This is achieved by appropriate immunohistochemicalanalysis; plasmacytoma, in contrast to lymphoma,does not express CD45 <strong>and</strong> immature B- <strong>and</strong>T-cell markers [351]. It also does not express antigenscharacteristic <strong>of</strong> malignant melanoma (i.e. S-100 protein,HMB-45 <strong>and</strong> melan-A), carcinoma (cytokeratins)<strong>and</strong> neuroendocrine neoplasms (i.e. synaptophysin,chromogranin).Plasmacytoma is radiosensitive <strong>and</strong> complete eradicationby radiation <strong>and</strong>/or surgery is potentially curative[183, 269, 270, 303, 378]. The prognosis is favourable,although <strong>the</strong> development <strong>of</strong> a plasma cell myelomamay occur in 15% <strong>of</strong> patients with extraosseous plasmacytoma[142].7.7.6.3 Primary Mucosal MelanomaICD-O:8720/3Primary malignant melanoma (MM) <strong>of</strong> <strong>the</strong> larynx is extremelyrare; less than 60 cases have been described in<strong>the</strong> literature. They represent 3.6 to 7.4% <strong>of</strong> all mucosalmelanomas <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck [6, 185, 212, 379].Primary laryngeal MM is more common in men,mostly in <strong>the</strong> 6th <strong>and</strong> 7th decades. It occurs primarilyin <strong>the</strong> supraglottic region <strong>and</strong> less <strong>of</strong>ten in <strong>the</strong> glottic region,but it has not yet been described in <strong>the</strong> subglottis.The symptoms vary according to <strong>the</strong> site <strong>of</strong> involvement<strong>and</strong> generally occur over a short period <strong>of</strong> time [379].Macroscopically, it may present as a polypoid, exophytic,nodular, sessile, or pedunculated lesion, with orwithout surface ulceration, varying in colour from blackor brown, to tan-grey or white.Microscopically, primary laryngeal MM is indistinguishablefrom MM <strong>of</strong> <strong>the</strong> skin <strong>and</strong> o<strong>the</strong>r mucous membranes.It may be composed <strong>of</strong> epi<strong>the</strong>lioid cells, spindlecells, or both. Nuclear <strong>and</strong> cellular pleomorphism, nuclearpseudoinclusions, mitoses <strong>and</strong> necroses are usuallyprominent.The diagnosis is based on histological examination,toge<strong>the</strong>r with special stainings for melanin, such as Fontana-Masson,<strong>and</strong> immunohistochemistry.The differential diagnosis must always include <strong>the</strong>possibility <strong>of</strong> a metastatic MM because in <strong>the</strong> larynx,metastatic MM is considerably more common than primaryMM [153]. Histologic features that favour <strong>the</strong> diagnosis<strong>of</strong> primary MM are junctional activity <strong>and</strong>/oran in situ component. However, as normal melanocytesare also normally present in <strong>the</strong> subepi<strong>the</strong>lial compartment,junctional changes are not required for <strong>the</strong> diagnosis<strong>of</strong> primary MM [379].Apart from metastatic MM, <strong>the</strong> differential diagnosisincludes carcinoma (especially spindle cell carcinoma),sarcoma <strong>and</strong> lymphoma. Positive staining for wellknownMM markers, such as S-100 protein, HMB-45,melan-A <strong>and</strong> vimentin, <strong>and</strong> negative staining for CD45,B- <strong>and</strong> T-cell markers, as well as markers <strong>of</strong> epi<strong>the</strong>lialdifferentiation (cytokeratins, epi<strong>the</strong>lial membrane antigen)is diagnostic for MM.The treatment <strong>of</strong> choice is complete surgical excision.The prognosis <strong>of</strong> primary laryngeal MM is poor, similarto primary mucosal malignant melanoma in general,with an average survival <strong>of</strong> less than 3.5 years [260,379].7.7.6.4 Metastases to <strong>the</strong> LarynxMetastases to <strong>the</strong> larynx from distant primary tumoursare uncommon, accounting for less than 0.5%<strong>of</strong> all laryngeal neoplasms. Metastases to <strong>the</strong> hypopharynx<strong>and</strong> trachea are even less common. The mostcommon source is malignant melanoma (Fig. 7.15),followed by renal cell carcinoma. O<strong>the</strong>r tumours withproven laryngeal metastases include cancer <strong>of</strong> <strong>the</strong>breast, lung, prostate, colon, stomach <strong>and</strong> ovary [24,74, 105, 154, 267, 296]. The rare occurrence <strong>of</strong> metastasesto <strong>the</strong> larynx seems to be related to <strong>the</strong> terminallocation <strong>of</strong> this organ in <strong>the</strong> lymphatic <strong>and</strong> vascularcirculation.Laryngeal metastases are most commonly located in<strong>the</strong> supraglottic <strong>and</strong> subglottic regions, presumably dueto <strong>the</strong>ir rich vascular supply [24, 133]. They can be dividedinto those located in <strong>the</strong> s<strong>of</strong>t tissue (metastasesfrom melanoma <strong>and</strong> renal cell carcinoma) <strong>and</strong> those locatedprimarily in <strong>the</strong> marrow spaces <strong>of</strong> <strong>the</strong> ossified laryngealcartilage (metastases from breast, prostate, <strong>and</strong>lung cancer).The signs <strong>and</strong> symptoms <strong>of</strong> metastatic laryngeal tumoursdo not differ from those <strong>of</strong> o<strong>the</strong>r laryngeal tumours<strong>and</strong> vary according to <strong>the</strong> site <strong>of</strong> involvement.Haemoptysis may be present, especially in highly vascularisedmetastatic renal cell carcinoma.


226 N. Gale · A. Cardesa · N. Zidar7Fig. 7.15. Metastastic malignant melanoma <strong>of</strong> <strong>the</strong> larynx. An ulceratedtumour composed <strong>of</strong> large, atypical epi<strong>the</strong>lioid cells withfocal melanin pigment. Primary tumour was located on <strong>the</strong> lef<strong>the</strong>elThe prognosis for patients with laryngeal metastasesis generally poor as laryngeal involvement is usually asign <strong>of</strong> dissemination in <strong>the</strong> terminal stage <strong>of</strong> <strong>the</strong> disease.In such patients, only palliative treatment is advised; laserendoscopic resection has been reported to be an excellenttool for relieving airway obstruction [267]. However,cases <strong>of</strong> isolated laryngeal metastases have been described,in which local excision <strong>and</strong>/or radiation <strong>the</strong>rapywas associated with prolonged survival [74, 105].References1. Abramson AL, Zielinski B (1984) Congenital laryngeal saccularcyst <strong>of</strong> <strong>the</strong> newborn. Laryngoscope 94:1580–15822. Absalom AR, Watts R, Kong A (1998) Airway obstructioncaused by rheumatoid cricoarytenoid arthritis. Lancet351:1099–11003. Alavi S, Namazie A, Calcaterra TC, Blackwell KE (1999)Gl<strong>and</strong>ular carcinoma <strong>of</strong> <strong>the</strong> larynx: <strong>the</strong> UCLA experience.Ann Otol Rhinol Laryngol 108:485–4894. Allen MS, Pettit JM, Sherblom JC (1991) Management <strong>of</strong>vocal nodules: a regional survey <strong>of</strong> otolaryngologists <strong>and</strong>speech-language pathologists. J Speech Hear Res 34:229–2355. Aluffi P, Prestinari A, Ramponi A, Castri M, Pia F (2000) Rosai-Dorfm<strong>and</strong>isease <strong>of</strong> <strong>the</strong> larynx. J Laryngol Otol 114:565–5676. Amin HM, Petruzzelli GJ, Husain AN, Nickol<strong>of</strong>fi BJ (2001)Primary malignant melanoma <strong>of</strong> <strong>the</strong> larynx. Arch PatholLab Med 125:271–2737. Amoils CP, Shindo ML (1996) Laryngotracheal manifestations<strong>of</strong> rhinoscleroma. Ann Otol Rhinol Laryngol 105:336–3408. Ansell S, Habermann TM, Hoyer JD, Strickler JG, Chen MG,McDonald TJ (1997) Primary laryngeal lymphoma. Laryngoscope107:1502–15069. Arber DA, Weiss LM, Chang KL (1998) Detection <strong>of</strong> Epstein-Barrvirus in inflammatory pseudotumor. Semin DiagnPathol 15:155–16010. Arens C, Glanz H, Kleinsasser O (1997) Clinical <strong>and</strong> morphologicalaspects <strong>of</strong> laryngeal cysts. Eur Arch Otorhinolaryngol254:430–43611. Asrar L, Facharzt EH, Haque I (1998) Rosai-Dorfman disease:presenting as isolated extranodal involvement <strong>of</strong> larynx.J Otolaryngol 27:85–8612. Assimakopoulos D, Patrikakos G (2002) The role <strong>of</strong> gastroesophagealreflux in <strong>the</strong> pathogenesis <strong>of</strong> laryngeal carcinoma.Am J Otolaryngol 23:351–35713. Austen KF (2001) Allergies, anaphylaxis <strong>and</strong> systemic mastocytosis.In: Braunwald E, Fauci AS, Kasper DL, Hauser SL,Longo DL, Jameson JL (eds) Harrison’s principles <strong>of</strong> internalmedicine, 15th edn, McGraw Hill, New York, pp 1913–192214. Bachman AL (1978) Benign, non-neoplastic conditions <strong>of</strong> <strong>the</strong>larynx <strong>and</strong> pharynx. Radiol Clin North Am 16:273–29015. Baker HL, Baker SR, McClatchey KD (1982) Manifestations<strong>and</strong> management <strong>of</strong> laryngoceles. <strong>Head</strong> <strong>Neck</strong> Surg 4:450–45616. Balestrieri F, Watson CB (1982) Intubation granuloma. OtolaryngolClin North Am 15:567–57917. Barnes C, Sexton M, Sizel<strong>and</strong> A, Tiedemann K, BerkowitzRG, Waters K (2001) Laryngo-pharyngeal carcinoma inchildhood. Int J Pediatr Otorhinolaryngol 61:83–8618. Barnes L (1991) Paraganglioma <strong>of</strong> <strong>the</strong> larynx. A critical review<strong>of</strong> <strong>the</strong> literature. ORL J Otorhinolaryngol Relat Spec53:220–23419. Barnes L (2001) Diseases <strong>of</strong> <strong>the</strong> larynx, hypopharynx, <strong>and</strong>esophagus. In: Barnes L (ed) Surgical pathology <strong>of</strong> <strong>the</strong> head<strong>and</strong> neck. Dekker, New York, pp127–23720. Barnes L, Ferlito A, Wenig BM (1997) Laryngeal paragangliomas.A review <strong>and</strong> report <strong>of</strong> a single case. J Laryngol Otol111:197–19821. Barnes L, Peel RL (1990) <strong>Head</strong> <strong>and</strong> neck pathology: a text/atlas<strong>of</strong> differential diagnosis. Igaku-Shoin, New York, pp 70–7722. Batsakis JG (1987) Laryngeal involvement by thyroid disease.Ann Otol Rhinol Laryngol 96:718–71923. Batsakis JG, El-Naggar AK, Luna MA (1992) Neuroendocrinetumors <strong>of</strong> <strong>the</strong> larynx. Ann Otol Rhinol Laryngol101:710–71424. Batsakis JG, Luna MA, Byers RM (1985) Metastases to <strong>the</strong>larynx. <strong>Head</strong> <strong>Neck</strong> Surg 7:458–46025. Batsakis JG, Luna MA, El-Naggar AK (1992) Nonsquamouscarcinomas <strong>of</strong> <strong>the</strong> larynx. Ann Otol Rhinol Laryngol102:1024–102626. Batsakis JG, Raymond AK (1988) Cartilage tumors <strong>of</strong> <strong>the</strong> larynx.South Med J 81:481–48427. Bengtsson M, Bengtsson A (1998) Cricoarytenoid arthritis– a cause <strong>of</strong> upper airway obstruction in <strong>the</strong> rheumatoid arthritispatient. Intensive Care Med 24:64328. Benjamin B, Croxson G (1985) Vocal cord granulomas. AnnOtol Rhinol Laryngol 94:538–54129. Benjamin B, Dalton C, Croxson G (1995) Laryngoscopic diagnosis<strong>of</strong> laryngeal sarcoid. Ann Otol Rhinol Laryngol104:529–55330. Benjamin B, Jacobson I, Eckstein R (1997) Idiopathic subglotticstenosis: diagnosis <strong>and</strong> endoscopic laser treatment.Ann Otol Rhinol Laryngol 106:770–77431. Benjamin B, Robb P, Clifford A, Eckstein R (1991) Giant Teflongranuloma <strong>of</strong> <strong>the</strong> larynx. <strong>Head</strong> <strong>Neck</strong> 13:453–45632. Bennett S, Bishop S, Lumpkin SM (1987) Phonatory characteristicsassociated with bilateral diffuse polypoid degeneration.Laryngoscope 97:446–45033. Bikhazi PH, Messina L, Mhatre AN, Goldstein JA, LalwaniAK (2000) Molecular pathogenesis in sporadic head <strong>and</strong>neck paraganglioma. Laryngoscope 110:1346–134834. Bilgic B, Mete Ö, Öztürk AS, Demiryont M, Keles N, BasaranM (2003) Synovial sarcoma: a rare tumor <strong>of</strong> <strong>the</strong> larynx.Pathol Oncol Res 9:242–245


Larynx <strong>and</strong> Hypopharynx Chapter 7 22735. Birzgalis AR, Farrington WT, O’Keefe L, Shaw J (1993) Localizedtracheopathia osteoplastica <strong>of</strong> <strong>the</strong> subglottis. J LaryngolOtol 107:352–35336. Bone RC, Biller HF, Irwin TM (1972) Intralaryngotrachealthyroid. Ann Otol Rhinol Laryngol 81:424–42837. Boring CC, Squires TS, Tong T, Montgomery S (1994) Cancerstatistics, 1994. CA Cancer J Clin 44:7–2638. Bosetti C, Talamini R, Levi F, Negri E, Franceschi S, AiroldiL, La Vecchia C (2002) Fried foods: a risk factor for laryngealcancer? Br J Cancer 87:1230–123339. Bossingham DH, Simpson FG (1996) Acute laryngeal obstructionin rheumatoid arthritis. BMJ 312:295–29640. Bower JS, Belen JE, Weg JG, Dantzker DR (1980) Manifestations<strong>and</strong> treatment <strong>of</strong> laryngeal sarcoidosis. Am Rev RespirDis 122:325–33241. Boyle JO, Coulthard SW, M<strong>and</strong>el RM (1991) Laryngeal involvementin disseminated coccidioidomycosis. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 117:433–43842. Bradley PJ, Kochaar A, Quraishi MS (1999) Pharyngealpouch carcinoma: real or imaginary risks? Ann Otol RhinolLaryngol 108:1027–103243. Brais B, Bouchard JP, Xie YG, Rochefort DL, Chretien N,Tome FM, Lafreniere RG, Rommens JM, Uyama E, NohiraO, Blumen S, Korczyn AD, Heutink P, Mathieu J, DuranceauA, Codere F, Fardeau M, Rouleau GA, Korcyn AD (1998)Short GCG expansions in <strong>the</strong> PABP2 gene cause oculopharyngealmuscular dystrophy. Nat Genet 18:164–16744. Br<strong>and</strong>wein M, Huvos A (1995) Laryngeal oncocytic cystadenomas.Eight cases <strong>and</strong> a literature review. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 121:1302–130545. Br<strong>and</strong>wein M, Levi G, Som P, Urken ML (1992) Paraganglioma<strong>of</strong> <strong>the</strong> inferior laryngeal paraganglia. A case report. ArchOtolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 118:994–99646. Br<strong>and</strong>wein M, Moore S, Som P, Biller H (1992) Laryngealchondrosarcomas: a clinicopathologic study <strong>of</strong> 11 cases includingtwo chondrosarcomas with additional malignantmesenchymal component. Laryngoscope 8:858–86747. Bridger MW, Jahn AF, van Nostr<strong>and</strong> AW (1980) Laryngealrheumatoid arthritis. Laryngoscope 90:296–30348. Brodsky L, Yoshpe N, Ruben RJ (1983) Clinical-pathologicalcorrelates <strong>of</strong> congenital subglottic hemangiomas. Ann OtolRhinol Laryngol 105:4–1849. Buley ID, Gatter KC, Kelly PM, Heryet A, Millard PR (1988)Granular cell tumours revisited. An immunohistological<strong>and</strong> ultrastructural study. Histopathology 12:263–27450. Canalis RF, Maxwell DS, Hemenway WG (1977) Laryngocele– an updated review. J Otolaryngol 6:191–19951. Carbone A, Passannante A, Gloghini A, Devaney KO, RinaldoA, Ferlito A (1999) Review <strong>of</strong> sinus histiocytosis withmassive lymphadenopathy (Rosai-Dorfman disease) <strong>of</strong> head<strong>and</strong> neck. Ann Otol Rhinol Laryngol 108:1095–110452. Carpenter RJ III, Banks PM, McDonald TJ, S<strong>and</strong>erson DR(1978) Sinus histiocytosis with massive lymphadenopathy(Rosai-Dorfman disease): report <strong>of</strong> a case with respiratorytract involvement. Laryngoscope 88:1963–196953. Cassano L, Lombardo P, Marchese-Ragona R, Pastore A(2000) Laryngopyocele: three new clinical cases <strong>and</strong> review<strong>of</strong> <strong>the</strong> literature. Eur Arch Otorhinolaryngol 257:507–51154. Cattaruzza MS, Maisonneuve P, Boyle P (1996) Epidemiology<strong>of</strong> laryngeal cancer. Oral Oncol Eur J Cancer 32B:293–30555. Cavalot AL, Preti G, Vione N, Nazionale G, Palonta F, FaddaGL (2001) Isolated primary non-Hodgkin’s malignant lymphoma<strong>of</strong> <strong>the</strong> larynx. J Laryngol Otol 115:324–32656. Cessna MH, Zhou H, Sanger WG, Perkins SL, Tripp S, PickeringD, Daines C, C<strong>of</strong>fin CM (2002) Expression <strong>of</strong> ALK1<strong>and</strong> p80 in inflammatory my<strong>of</strong>ibroblastic tumor <strong>and</strong> its mesenchymalmimics: a study <strong>of</strong> 135 cases. Mod Pathol 15:931–93857. Chiu LD, Rasgon BM (1996) Laryngeal chondroma: a benignprocess with long-term clinical implications. Ear NoseThroat J 75:540–542; 544–54958. Cho MS, Kim ES, Kim HJ, Yang WI (1997) Kimura’s disease<strong>of</strong> <strong>the</strong> epiglottis. Histopathology 30:592–59459. Chow LT, Chow WH, Shum BS (1993) Fatal massive upper respiratorytract haemorrhage: an unusual complication <strong>of</strong> localizedamyloidosis <strong>of</strong> <strong>the</strong> larynx. J Laryngol Otol 107:51–5360. Chu L, Gussack GS, Orr JB, Hood D (1994) Neonatal laryngoceles.A cause for airway obstruction. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg120:454–45861. Cleary KR, Batsakis JG (1995) Mycobacterial disease <strong>of</strong> <strong>the</strong>head <strong>and</strong> neck: current perspective. Ann Otol Rhinol Laryngol104:830–83362. Close LG, Merkel M, Burns DK, Deaton CW Jr, Schaefer SD(1987) Asymptomatic laryngocele: incidence <strong>and</strong> associationwith laryngeal cancer. Ann Otol Rhinol Laryngol 96:393–39963. C<strong>of</strong>fin CM, Humphrey PA, Dehner LP (1998) Extrapulmonaryinflammatory my<strong>of</strong>ibroblastic tumor: a clinical <strong>and</strong>pathological survey. Semin Diagn Pathol 15:85–10164. C<strong>of</strong>fin CM, Watterson J, Priest JR, Dehner LP (1995) Extrapulmonaryinflammatory my<strong>of</strong>ibroblastic tumor (inflammatorypseudotumor). A clinicopathologic <strong>and</strong> immunohistochemicalstudy <strong>of</strong> 84 cases. Am J Surg Pathol 19:859–87265. Corsi A, Ci<strong>of</strong>alo A, Leonardi M, Zambetti G, Bosman C(1997) Recurrent inflammatory my<strong>of</strong>ibroblastic tumor <strong>of</strong> <strong>the</strong>glottis mimicking malignancy. Am J Otolaryngol 18:121–12666. Courteney-Harris RG, Goddard MJ (1992) Sinus histiocytosiswith massive lymphadenopathy (Rosai-Dorfman disease):a rare case <strong>of</strong> subglottic narrowing. J Laryngol Otol 106:61–6267. Cuzzourt JC, Pezold JC, Dunn CW (2002) Typical carcinoidtumor <strong>of</strong> <strong>the</strong> larynx occurring with otalgia. Ear Nose Throat81:40–4368. Damiani JM, Levine HL (1979) Relapsing polychondritis: report<strong>of</strong> ten cases. Laryngoscope 89:929–94669. Danish HM, Meleca RJ, Dworkin JP, Abbarah TR (1998) Laryngealobstructing saccular cysts: a review <strong>of</strong> this disease<strong>and</strong> treatment approach emphasizing complete endoscopiccarbon dioxide laser excision. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong>Surg 124:593–59670. De Bree R, Mahieu HF, Ossenkoppele GJ, van der Valk P(1998) Malignant lymphoma <strong>of</strong> mucosa-associated lymphoidtissue in <strong>the</strong> larynx. Eur Arch Otorhinolaryngol 255:368–37071. De Carvalho MB (1998) Quantitative analysis <strong>of</strong> <strong>the</strong> extent<strong>of</strong> extracapsular invasion <strong>and</strong> its prognostic significance: aprospective study <strong>of</strong> 170 cases <strong>of</strong> carcinoma <strong>of</strong> <strong>the</strong> larynx <strong>and</strong>hypopharynx. <strong>Head</strong> <strong>Neck</strong> 20:16–2172. Dean CM, Satal<strong>of</strong>f RT, Hawkshaw MJ, Pribikin E (2002) Laryngealsarcoidosis. J Voice 16:283–28873. Dedo HH, Catten MD (2001) Idiopathic progressive subglotticstenosis: findings <strong>and</strong> treatment in 52 patients. Ann OtolRhinol Laryngol 110:305–31174. Dee SL, Eshghi M, Otto CS (2000) Laryngeal metastasis7 years after radical nephrectomy. Arch Pathol Lab Med124:1833–183475. Deeb ZE (1997) Acute supraglottitis in adults: early indicators<strong>of</strong> airway obstruction. Am J Otolaryngol 18:112–11576. Dehner LP (2000) The enigmatic inflammatory pseudotumours:<strong>the</strong> current state <strong>of</strong> our underst<strong>and</strong>ing, or misunderst<strong>and</strong>ing.J Pathol 192:277–27977. Delahunty JE (1972) Acid laryngitis. J Laryngol Otol 86:335–34278. Delap TG, Lavy JA, Alusi G, Quiney RE (1997) Tuberculosispresenting as a laryngeal tumour. J Infect 34:139–14179. DeRienzo DP, Greenberg SD, Fraire AE (1991) Carcinoma <strong>of</strong><strong>the</strong> larynx. Changing incidence in women. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 117:681–68480. DeSanto LW, Devine KD, Weil<strong>and</strong> LH (1970) Cysts <strong>of</strong> <strong>the</strong>larynx – classification. Laryngoscope 80:145–17681. Deutsch ES, Zalzal GH (1991) Quincke’s edema, revisited.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 117:100–10282. Devaney KO, Ferlito A, Hunter BC, Devaney SL, Rinaldo A(1998) Wegener’s granulomatosis <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. AnnOtol Rhinol Laryngol 107:439–445


228 N. Gale · A. Cardesa · N. Zidar783. Devaney KO, Ferlito A, Silver CE (1995) Cartilaginous tumors<strong>of</strong> <strong>the</strong> larynx. Ann Otol Rhinol Laryngol 104:251–25584. Dhingra JK, Aqel NM, McEwen J, Bleach NR (1995) Multipleoncocytic cysts <strong>of</strong> <strong>the</strong> larynx. J Laryngol Otol 109:1226–122885. Dikkers FG, Nikkels PG (1995) Benign lesions <strong>of</strong> <strong>the</strong> vocalfolds: histopathology <strong>and</strong> phonotrauma. Ann Otol RhinolLaryngol 104:698–70386. Donovan DT, Conley J (1983) Adenoid cystic carcinoma <strong>of</strong><strong>the</strong> subglottic region. Ann Otol Rhinol Laryngol 92:491–49587. Dort JC, Frohlich AM, Tate RB (1994) Acute epiglottitis inadults: diagnosis <strong>and</strong> treatment in 43 patients. J Otolaryngol23:281–28588. Dubey SP, Banerjee S, Ghosh LM, Roy S (1997) Benign pleomorphicadenoma <strong>of</strong> <strong>the</strong> larynx: report <strong>of</strong> a case <strong>and</strong> review<strong>and</strong> analysis <strong>of</strong> 20 additional cases in <strong>the</strong> literature. Ear NoseThroat J 76:548-550; 552; 554–55789. Dumich PS, Neel HB III (1983) Blastomycosis <strong>of</strong> <strong>the</strong> larynx.Laryngoscope 93:1266–127090. Ebo DG, Stevens WJ (2000) Hereditary angioneurotic edema:review <strong>of</strong> <strong>the</strong> literature. Acta Clin Belg 55:22–2991. El-Naggar AK, Batsakis JG (1991) Carcinoid tumor <strong>of</strong> <strong>the</strong>larynx. A critical review <strong>of</strong> <strong>the</strong> literature. ORL OtorhinolaryngolRelat Spec 53:188–19392. El-Naggar AK, Batsakis JG, Luna MA (1992) Neuroendocrinetumors <strong>of</strong> larynx. Ann Otol Rhinol Laryngol 101:710–71493. Eng J, Sabanathan S (1991) Airway complications in relapsingpolychondritis. Ann Thorac Surg 51:686–69294. Epstein SS, Winston P, Friedmann I, Ormerod FC (1957) Thevocal cord polyp. J Laryngol Otol 71:673–68895. Erb N, Pace AV, Delamere JP, Kitas GD (2001) Dysphagia <strong>and</strong>stridor caused by laryngeal rheumatoid arthritis. Rheumatology40:952–95396. Ereno C, Lopez JI, Gr<strong>and</strong>e J, Santaolalla F, Bilbao FJ (2001)Inflammatory my<strong>of</strong>ibroblastic tumour <strong>of</strong> <strong>the</strong> larynx. J LaryngolOtol 115:856–85897. Ereno C, Lopez JI, Sanchez JM (1997) Atypical carcinoid <strong>of</strong>larynx: presentation with scalp metastases. J Laryngol Otol111:89–9198. Esteve J, Riboli E, Pequignot G, Terracini B, Merletti F, CrosignaniP, Ascunce N, Zubiri L, Blanchet F, Raymond L,Repetto F, Tuyns AJ (1996) Diet <strong>and</strong> cancers <strong>of</strong> <strong>the</strong> larynx<strong>and</strong> hypopharynx: <strong>the</strong> IARC multi-center study in southwesternEurope. Cancer Causes Control 7:240–25299. Evans HL, Ayala AG, Romsdahl MM (1977) Prognostic factorsin chondrosarcoma <strong>of</strong> bone: a clinicopathologic analysiswith emphasis on histologic grading. Cancer 40:818–831100. Fajardo-Dolci G, Chavolla R, Lamadrid-Bautista E, Rizo-AlvarezJ (1999) Laryngeal scleroma. J Otolaryngol 28:229–231101. Farchi G (1992) Estimation <strong>of</strong> <strong>the</strong> impact <strong>of</strong> cigarette smokingreduction on mortality. Ann Ist Super Sanita 28:147–53102. Fauci AS (1998) The vasculitis syndromes. In: Fauci AS,Braunwald E, Isselbacher KJ, Wilson JD, Martin JB, KasperDL, Hauser SL, Longo DL (eds) Harrison’s principles <strong>of</strong> internalmedicine, 14th edn. McGraw-Hill, New York, pp 1910–1922103. Fechner RE, Mills SE (1997) Larynx <strong>and</strong> pharynx. In: SterrnbergSS (ed) Histology for pathologists. Lippincott-Raven,Philadelphia, pp 391–403104. Feder RJ, Michell MJ (1984) Hyperfunctional, hyperacidic,<strong>and</strong> intubation granulomas. Arch Otolaryngol 110:582–584105. Ferlito A (1993) Secondary neoplasms. In: Ferlito A (ed) Neoplasms<strong>of</strong> <strong>the</strong> larynx. Churchill Livingstone, Edinburgh, pp349–360106. Ferlito A, Barnes L, Rinaldo A, Gnepp DR, Milroy CM (1998)A review <strong>of</strong> neuroendocrine neoplasms <strong>of</strong> <strong>the</strong> larynx: updateon diagnosis <strong>and</strong> treatment. J Laryngol Otol 112:827–834107. Ferlito A, Barnes L, Wenig BM (1994) Identification, classification,treatment, <strong>and</strong> prognosis <strong>of</strong> laryngeal paraganglioma.Review <strong>of</strong> <strong>the</strong> literature <strong>and</strong> eight new cases. Ann OtolRhinol Laryngol 103:525–536108. Ferlito A, Friedmann I (1993) Neuroendocrine neoplasms.In: Ferlito A (ed) Neoplasms <strong>of</strong> <strong>the</strong> larynx. Churchill Livingstone,Edinburgh, pp 169–205109. Ferlito A, Gale N, Hvala A (1981) Laryngeal salivary ductcarcinoma: a light <strong>and</strong> electron microscopic study. J LaryngolOtol 95:731–738110. Ferlito A, Milroy CM, Wenig BM, Barnes L, Silver CE (1995)Laryngeal paraganglioma versus atypical carcinoid tumor.Ann Otol Rhinol Laryngol 104:78–83111. Ferlito A, Recher G (1981) Oncocytic lesions <strong>of</strong> <strong>the</strong> larynx.Arch Otorhinolaryngol 232:107–115112. Ferlito A, Recher G (1985) Chondrometaplasia <strong>of</strong> <strong>the</strong> larynx.ORL J Otorhinolaryngol Relat Spec 47:174–177113. Ferlito A, Rinaldo A (2000) The pathology <strong>and</strong> management<strong>of</strong> subglottic cancer. Eur Arch Otorhinolaryngol 257:168–173114. Ferlito A, Rinaldo A (2003) Small cell neuroendocrine carcinoma<strong>of</strong> <strong>the</strong> larynx: a preventable <strong>and</strong> frustrating diseasewith a highly aggressive lethal behavior. ORL J OtorhinolaryngolRelat Spec 65:131–133115. Ferlito A, Rinaldo A, Devaney KO, Devaney SL, Milroy CM(1998) Impact <strong>of</strong> phenotype on treatment <strong>and</strong> prognosis <strong>of</strong>laryngeal malignancies. J Laryngol Otol 112:710–714116. Ferlito A, Rinaldo A, Devaney KO, MacLennan K, Myers JN,Petruzzelli GJ, Shaha AR, Genden EM, Johnson JT, de CarvalhoM, Myers EN (2002) Prognostic significance <strong>of</strong> microscopic<strong>and</strong> macroscopic extracapsular spread from metastatictumor in <strong>the</strong> cervical lymph nodes. Oral Oncol 38:747–751117. Ferlito A, Shaha AR, Silver CE, Rinaldo A, Mondin V (2001)Incidence <strong>and</strong> sites <strong>of</strong> distant metastases from head <strong>and</strong> neckcancer. ORL J Otorhinolaryngol Relat Spec 63:202–207118. Foucar E, Rosai J, Dorfman R (1990) Sinus histiocytosis withmassive lymphadenopathy (Rosai-Dorfman disease): review<strong>of</strong> <strong>the</strong> entity. Semin Diagn Pathol 7:19–73119. Foulsham CK II, Snyder GG III, Carpenter RJ III (1981) Papillarycystadenoma lymphomatosum <strong>of</strong> <strong>the</strong> larynx. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 89:960–964120. Franceschi S (2003) Fibre intake <strong>and</strong> laryngeal cancer risk.Ann Oncol 14:162–167121. Frantz TD, Rasgon BM, Quesenberry CP Jr (1994) Acute epiglottitisin adults. Analysis <strong>of</strong> 129 cases. JAMA 272:1358–1360122. Freije JE, Beatty TW, Campbell BH, Woodson BT, Schultz CJ,Toohill RJ (1996) Carcinoma <strong>of</strong> <strong>the</strong> larynx in patients withgastroesophageal reflux. Am J Otolaryngol 17:386–390123. Frenzel H, Schmitt-Graff A, Seitz RJ (1985) An immunohistochemical<strong>and</strong> electron-microscopic study <strong>of</strong> vascular endo<strong>the</strong>lialcells in vocal fold polyps. Arch Otorhinolaryngol241:271–278124. Gale N, Zidar N, Podboj J, Volavšek M, Luzar B (2003) Inflammatorymy<strong>of</strong>ibroblastic tumour <strong>of</strong> paranasal sinuseswith fatal outcome: reactive lesion or tumour? J Clin Pathol56:715–717125. Galli J, Nardi C, Contucci AM, Cadoni G, Lauriola L, FantoniM (2002) Atypical isolated epiglottic tuberculosis: acase report <strong>and</strong> a review <strong>of</strong> <strong>the</strong> literature. Am J Otolaryngol23:237–240126. Gallo O, Bianchi S, Giannini A, Boccuzzi S, Calzolari A, Fini-Storchi O (1994) Lack <strong>of</strong> detection <strong>of</strong> human papillomavirus(HPV) in transformed laryngeal keratoses by in situ hybridization(ISH) technique. Acta Otolaryngol 114:213–217127. Gallo A, de Vicentiis M, Della Rocca C, Moi R, Simonelli M,Minni A, Shaha AR (2001) Evolution <strong>of</strong> precancerous lesions:a clinicopathologic study with long-term follow-up on 259patients. <strong>Head</strong> <strong>Neck</strong> 23:42–47128. Garcia RE, Gannon FH, Thompson LDR (2002) Dedifferentiatedchondrosarcoma <strong>of</strong> <strong>the</strong> larynx: a report <strong>of</strong> two cases <strong>and</strong>review <strong>of</strong> <strong>the</strong> literature. Laryngoscope 112:1015–1018129. Gervais M, Dorion D (2003) Quality <strong>of</strong> life following surgicaltreatment <strong>of</strong> oculopharyngeal syndrome. J Otolaryngol32:1–5130. Geterud A (1991) Rheumatoid arthritis in <strong>the</strong> larynx. Sc<strong>and</strong>J Rheumatol 20:215131. Gillmore JD, Hawkins PN, Pepys MB (1997) Amyloidosis: areview <strong>of</strong> recent diagnostic <strong>and</strong> <strong>the</strong>rapeutic developments. BrJ Haematol 99:245–256


Larynx <strong>and</strong> Hypopharynx Chapter 7 229132. Giudice M, Piazza C, Foccoli P, Toninelli C, Cavaliere S, PerettiG (2003) Idiopathic subglottic stenosis: management byendoscopic <strong>and</strong> open-neck surgery in a series <strong>of</strong> 30 patients.Eur Arch Otorhinolaryngol 260:235–238133. Glanz H, Kleinsasser O (1978) Metastasen im Kehlkopf.HNO 26:163–167134. Glaubiger DL, Casler JD, Garrett WL, Yuo HS, Lilis-HearnePK (1991) Chondrosarcoma <strong>of</strong> <strong>the</strong> larynx after radiationtreatment for vocal cord cancer. Cancer 68:1828–1831135. Gnepp DR (1991) Small cell neuroendocrine carcinoma <strong>of</strong><strong>the</strong> larynx: a critical review <strong>of</strong> <strong>the</strong> literature. ORL J OtorhinolaryngolRelat Spec 53:210–219136. Gonzales-Lois C, Ibarrola C, Ballestin C, Martanez-Tello FJ(2002) Dedifferentiated liposarcoma <strong>of</strong> <strong>the</strong> pyriform sinus:report <strong>of</strong> a case <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Int J Surg Pathol10:75–79137. Goutas N, Simopoulou S, Papazoglou K, Agapitos E (1994) Afatal case <strong>of</strong> diph<strong>the</strong>ria. Pediatr Pathol 14:391–395138. Gray SD, Hammond E, Hanson DF (1995) Benign pathologicresponses <strong>of</strong> <strong>the</strong> larynx. Ann Otol Rhinol Laryngol 104:13–18139. Gregg CM, Wiatrak BJ, Koopmann CF Jr (1995) Managementoptions for infantile subglottic hemangioma. Am JOtolaryngol 16:409–414140. Gregor RT, L<strong>of</strong>tus B, Cohen P, Balm AJ, Hilgers FJ (1994)Saccular mucocele in association with laryngeal cancer. AnnOtol Rhinol Laryngol 103:732–736141. Gripp S, Pape H, Scmitt G (1998) Chondrosarcoma <strong>of</strong> <strong>the</strong> larynx– <strong>the</strong> role <strong>of</strong> radio<strong>the</strong>rapy revisited – case report <strong>and</strong> review<strong>of</strong> <strong>the</strong> literature. Cancer 82:108–115142. Grogan TM, Van Camp B, Kyle RA, Muller-Hermelink HK,Harris NL (2001) Plasma cell neoplasms. In: Jaffe ES, HarrisNL, Stein H, Vardiman JW (eds) Tumours <strong>of</strong> haematopoietic<strong>and</strong> lymphoid tissues. IARC Press. Lyon, pp 142–156143. Guney E, Yigitbasi OG, Balkanli S, Canoz OM (2002) Postirradiationmalignant fibrous histiocytoma <strong>of</strong> <strong>the</strong> larynx: acase report. Am J Otolaryngol 23:293–296144. Gunkel AR, Thurner KH, Kanonier G, Sprinzl GM, ThumfartWF (1996) Angioneurotic edema as a reaction to angiotensin-convertingenzyme inhibitors. Am J Otolaryngol17:87–91145. Gupta OP, Jain RK, Tripathi PP, Gupta S (1984) Leprosy <strong>of</strong><strong>the</strong> larynx: a clinicopathological study. Int J Lepr O<strong>the</strong>r MycobactDis 52:171–175146. Guttenplan MD, Hendrix RA, Townsend MJ, Balsara G(1991) Laryngeal manifestations <strong>of</strong> gout. Ann Otol RhinolLaryngol 100:899–902147. Hakky M, Kolbusz R, Reyes CV (1989) Chondrosarcoma <strong>of</strong><strong>the</strong> larynx. Ear Nose Throat 68:60–62148. Härmä RA, Suurkari S (1977) Tracheopathia chondro-osteoplastica.A clinical study <strong>of</strong> thirty cases. Acta Otolaryngol84:118–123149. Harrison DFN (1971) The pathology <strong>and</strong> management <strong>of</strong>subglottic cancer. Ann Otol Rhinol Laryngol 80:6–12150. Hébert PC, Ducic Y, Boisvert D, Lamo<strong>the</strong> A (1998) Adult epiglottitisin a Canadian setting. Laryngoscope 108:64–69151. Helliwell TR (2003) Evidence based pathology: squamouscarcinoma <strong>of</strong> <strong>the</strong> hypopharynx. J Clin Pathol 56:81–85152. Heman-Ackah YD, Remley KB, Goding GS Jr (1999) A newrole for magnetic resonance imaging in <strong>the</strong> diagnosis <strong>of</strong> laryngealrelapsing polychondritis. <strong>Head</strong> <strong>Neck</strong> 21:484–489153. Henderson LT, Robbins KT, Weitzner S (1986) Upper aerodigestivetract metastases in disseminated malignant melanoma.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 112:659–663154. Hilger AW, Prichard AJ, Jones T (1998) Adenocarcinoma <strong>of</strong><strong>the</strong> larynx – a distant metastasis from a rectal primary. J LaryngolOtol 112:199–201155. Hill ME, Creed GA, McMullan TF, Tyers AG, Hilton-JonesD, Robinson DO, Hammans SR (2001) Oculopharyngealmuscular dystrophy: phenotypic <strong>and</strong> genotypic studies in aUK population. Brain 124:522–526156. Hill MJ, Taylor CL, Scott GB (1980) Chondromatous metaplasiain <strong>the</strong> human larynx. Histopathology 4:205–214157. Hirabayashi H, Koshii K, Uno K, Ohgaki H, Nakasone Y, FujisawaT, Syouno N, Hinohara T, Hirabayashi K (1991) Extracapsularspread <strong>of</strong> squamous cell carcinoma in neck lymphnodes: prognostic factor <strong>of</strong> laryngeal cancer. Laryngoscope101:501–506158. Hirano M, Sato K (1993) Histological color atlas <strong>of</strong> <strong>the</strong> humanlarynx. Singular, San Diego, pp 1–112159. Hočevar-Boltežar I, Zidar N, Žargi M, Župevc A, Lestan B,Andoljšek D (2000) Amyloidosis <strong>of</strong> <strong>the</strong> larynx. Wien KlinWochenschr 112:732–734160. H<strong>of</strong>fman GS, Kerr GS, Leavitt RY, Hallahan CW, LebovicsRS, Travis WD, Rottem M, Fauci AS (1992) Wegener granulomatosis:an analysis <strong>of</strong> 158 patients. Ann Intern Med116:488–498161. Holinger LD, Barnes DR, Smid LJ, Holinger PH (1978) Laryngocele<strong>and</strong> saccular cysts. Ann Otol Rhinol Laryngol87:675–685162. Holl<strong>and</strong> RS, Abaza N, Balsara G, Lesser R (1998) Granularcell tumor <strong>of</strong> <strong>the</strong> larynx in a six-year-old child: case report<strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Ear Nose Throat J 77:652–654;656; 658163. Horny HP, Kaiserling E (1995) Involvement <strong>of</strong> <strong>the</strong> larynx byhemopoietic neoplasms. An investigation <strong>of</strong> autopsy cases<strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Pathol Res Pract 191:130–138164. Hughes RA, Berry CL, Seifert M, Less<strong>of</strong> MH (1972) Relapsingpolychondritis. Three cases with a clinico-pathologicalstudy <strong>and</strong> literature review. Q J Med 41:363–380165. Hussong JW, Brown M, Perkins SL, Dehner LP, C<strong>of</strong>fi n CM(1999) Comparison <strong>of</strong> DNA ploidy, histologic, <strong>and</strong> immunohistochemicalfindings with clinical outcome in inflammatorymy<strong>of</strong>ibroblastic tumors. Mod Pathol 12:279–286166. Hyams VJ, Rabuzzi DD (1970) Cartilaginous tumors <strong>of</strong> <strong>the</strong>larynx. Laryngoscope 80:755–767167. Hytiroglou P, Br<strong>and</strong>wein MS, Strauchen JA, Mirante JP,Urken ML, Biller HF (1992) Inflammatory pseudotumor <strong>of</strong><strong>the</strong> parapharyngeal space: case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.<strong>Head</strong> <strong>Neck</strong> 14:230–234168. Ibrahim R, Bird DJ, Sieler MW (1991) Malignant myoepi<strong>the</strong>lioma<strong>of</strong> <strong>the</strong> larynx with massive metastatic spread to <strong>the</strong> liver– an ultrastructural <strong>and</strong> immunocytochemical study. UltrastructPathol 15:69–76169. Imauchi Y, Ito K, Takasago E, Nibu KI, Sugasawa M, IchimuraK (2002) Stomal recurrence after total laryngectomy forsquamous cell carcinoma <strong>of</strong> <strong>the</strong> larynx. Otolaryngol <strong>Head</strong><strong>Neck</strong> Surg 126:63–66170. Inglis AF Jr (1993) Herpes simplex virus infection. A rarecause <strong>of</strong> prolonged croup. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg119:551–552171. Isaacson JE, Frable MA (1996) Cryptococcosis <strong>of</strong> <strong>the</strong> larynx.Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 114:106–109172. Jennette JC (1994) Pathogenic potential <strong>of</strong> anti-neutrophilcytoplasmic autoantibodies. Lab Invest 70:135–137173. Jennings CR, Jones NS, Dugar J, Powell RJ, Lowe J (1998) Wegener’sgranulomatosis – a review <strong>of</strong> diagnosis <strong>and</strong> treatmentin 53 subjects. Rhinology 36:188–191174. Jindal JR, Milbrath MM, Shaker R, Hogan WJ, Toohill RJ(1994) Gastroesophageal reflux disease as a likely cause <strong>of</strong>“idiopathic” subglottic stenosis. Ann Otol Rhinol 103:186–191175. Johns MM (2003) Update on <strong>the</strong> etiology, diagnosis, <strong>and</strong>treatment <strong>of</strong> vocal fold nodules, polyps, <strong>and</strong> cysts. Curr OpinOtolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 11:456–461176. Kamal SA, Othman EO (1998) Granular cell tumour <strong>of</strong> <strong>the</strong>larynx. J Laryngol Otol 112:83–85177. Kambič V, Gale N (1995) Epi<strong>the</strong>lial hyperplastic lesions <strong>of</strong><strong>the</strong> larynx. Elsevier, Amsterdam, pp 1–265178. Kambič V, Gale N, Radšel Z (1982) Warty dyskeratoma <strong>of</strong> <strong>the</strong>vocal cord. First reported case. Arch Otolaryngol 108:385–387179. Kambič V, Gale N, Radšel Z (1989) Anatomical markers <strong>of</strong>Reinke’s space <strong>and</strong> <strong>the</strong> etiopathogenesis <strong>of</strong> Reinke edema.Laryngorhinootologie 68:231–235180. Kambič V, Radšel Z (1984) Acid posterior laryngitis. Aetiology,histology, diagnosis <strong>and</strong> treatment. J Laryngol Otol98:1237–1240


230 N. Gale · A. Cardesa · N. Zidar7181. Kambič V, Radšel Z, Žargi M, Ačko M (1981) Vocal cord polyps:incidence, histology <strong>and</strong> pathogenesis. J Laryngol Otol95:609–618182. Kambič V, Žargi M, Gale N (1989) Laryngeal chondrosarcoma:is conservative surgery adequate treatment? J LaryngolOtol 103:970–972183. Kamijo T, Inagi K, Nakajima M, Motoori T, Tadokoro K,Nishiyam S (2002) A case <strong>of</strong> extramedullary plasmacytoma<strong>of</strong> <strong>the</strong> larynx. Acta Otolaryngol Suppl 547:104–107184. K<strong>and</strong>iloros DC, Nikolopoulos TP, Ferekidis EA, TsangaroulakisA, Yiotakis JE, Davilis D, Adamopoulos GK (1997) Laryngealtuberculosis at <strong>the</strong> end <strong>of</strong> <strong>the</strong> 20th century. J LaryngolOtol 111:619–621185. Karagiannidis K, Noussios G, Sakellariou T (1998) Primarylaryngeal melanoma. J Otolaryngol 27:104–106186. Karlikaya C, Yuksel M, Kilicli S, C<strong>and</strong>an L (2000) Tracheobronchopathiaosteochondroplastica. Respirology 5:377–380187. Kawaida M, Fukuda H, Kohno N (2000) Granular cell tumorsarising nearly simultaneously in <strong>the</strong> larynx <strong>and</strong> subcutaneouscervical region. Ear Nose Throat J 79:162–166188. Kendall CH, Johnston MN (1998) Pseudo-malignant laryngealnodule (inflammatory my<strong>of</strong>ibroblastic tumour). Histopathology32:286–287189. Kenny TJ, Werkhaven J, Netterville JL (2000) Sarcoidosis<strong>of</strong> <strong>the</strong> pediatric larynx. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg126:536–539190. Kershisnik M, Batsakis JG, Mackay B (1994) Granular celltumors. Ann Otol Rhinol Laryngol 103:416–419191. Kienast U, Neumann HJ, Stintz S (1997) Isolated carcinoid <strong>of</strong><strong>the</strong> larynx – a rare neuroendocrine tumor <strong>of</strong> <strong>the</strong> upper airways.Otorhinolaryngol Nova 7:286–287192. Kirchner JA, Som JL (1971) Clinical significance <strong>of</strong> fixed vocalcord. Laryngoscope 81:1029–1044193. Kleidermacher P, Mastros NP, Miller FR, Jones EW, WoodBG (1995) Pathologic quiz case 2. Oncocytic cyst <strong>of</strong> <strong>the</strong> larynx.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 121:1430–1431; 1433194. Kleinsasser O (1982) Pathogenesis <strong>of</strong> vocal cord polyps. AnnOtol Rhinol Laryngol 91:378–381195. Kleinsasser O (1988) Tumors <strong>of</strong> <strong>the</strong> larynx <strong>and</strong> hypopharynx.Thieme, Stuttgart196. Kontzoglou G, Triaridis S, Noussios G, Valeri R, Nanas C(2002) Subglottic hemangioma treated with interferon alpha2A. Acta Otorhinolaryngol Belg 56:83–85197. Krespi YP, Mitrani M, Husain S, Meltzer CJ (1987) Treatment<strong>of</strong> laryngeal sarcoidosis with intralesional steroid injection.Ann Otol Rhinol Laryngol 96:713–715198. Kurtin PJ, Bonin DM (1994) Immunohistochemical demonstration<strong>of</strong> <strong>the</strong> lysosome-associated glycoprotein CD68 (KP-1) in granular cell tumors <strong>and</strong> schwannomas. Hum Pathol25:1172–1178199. Kutta H, Steven P, Tillmann BN, Tsokos M, Paulsen FP(2003) Region-specific immunological response <strong>of</strong> <strong>the</strong> differentlaryngeal compartments: significance <strong>of</strong> larynx-associatedlymphoid tissue. Cell Tissue Res 311:365–371200. La Veccia C, Negri E, D’Avanzo B, Franceschi S, Decarli A,Boyle P (1990) Dietary indicators <strong>of</strong> laryngeal cancer risk.Cancer Res 50:4497–4500201. Lack EE, Worsham GF, Callihan MD, Crawford BE, KlappenbachS, Rowden G, Chun B (1980) Granular cell tumor: aclinicopathologic study <strong>of</strong> 110 patients. J Surg Oncol 13:301–316202. Lan MY, Guo YC, Chu PY, Ho DM, Chang SY (2001) <strong>Pathology</strong>quiz case 2: leiomyosarcoma <strong>of</strong> <strong>the</strong> larynx. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 127:1503–1505203. Lang WS (1965) Diph<strong>the</strong>ria at <strong>the</strong> present time. Laryngoscope75:1092–1102204. Larsen WJ (1997) Human embryology, 2nd edn. ChurchillLivingstone, New York, pp 347–374205. Lassaletta L, Alonso S, Ballestin C, Martinez-Tello FJ, Alvarez-VicentJJ (1999) Immunoreactivity in granular cell tumours<strong>of</strong> <strong>the</strong> larynx. Auris Nasus Larynx 26:305–310206. Lassaletta L, Alonso S, Granell J, Ballestin C, Serrano A, Alvarez-VincentJJ (1998) Synchronous glottic granular cell tumor<strong>and</strong> subglottic spindle cell carcinoma. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 124:1031–1034207. Leighton SE, Gallimore AP (1994) Extranodal sinus histiocytosiswith massive lymphadenopathy affecting <strong>the</strong> subglottis<strong>and</strong> trachea. Histopathology 24:393–394208. Lerner DM, Deeb Z (1993) Acute upper airway obstructionresulting from systemic diseases. South Med J 86:623–627209. Lewis JE, Olsen KD, Inwards CY (1997) Cartilaginous tumors<strong>of</strong> <strong>the</strong> larynx: clinicopathologic review <strong>of</strong> 47 cases. AnnOtol Rhinol Laryngol 106:94–100210. Lewy RB, Millet D (1978) Immediate local tissue reactions toTeflon vocal cord implants. Laryngoscope 88:1339–1342211. Lie ES, Karlsen F, Holm R (1996) Presence <strong>of</strong> human papillomavirusin squamous cell laryngeal carcinomas. A study <strong>of</strong>thirty-nine cases using polymerase chain reaction <strong>and</strong> in situhybridization. Acta Otolaryngol 116:900–905212. Lin SY, Hsu CY, Jan YJ (2001) Primary laryngeal melanoma.Otolaryngol <strong>Head</strong> <strong>Neck</strong> 125:569–570213. Lindeberg H, Krogdahl A (1999) Laryngeal cancer <strong>and</strong> humanpapillomavirus: HPV is absent in <strong>the</strong> majority <strong>of</strong> laryngealcarcinomas. Cancer Lett 146:9–13214. Loos BM, Wieneke JA, Thompson LDR (2001) Laryngeal angiosarcoma:clinicopathologic study <strong>of</strong> five cases with a review<strong>of</strong> <strong>the</strong> literature. Laryngoscope 111:1197–1202215. Luna MA, Pineda-Daboin K (2001) Upper aerodigestivetract. In: Henson D, Albores-Saavedra J <strong>Pathology</strong> <strong>of</strong> incipientneoplasia, 3rd edn. Oxford University Press, New York,pp 57–85216. Lundgren J, Ol<strong>of</strong>sson J, Hellquist H (1982) Oncocytic lesions<strong>of</strong> <strong>the</strong> larynx. Acta Otolaryngol 94:335–344217. Luzar B, Gale N, Klopčič U, Fischinger J (2000) Laryngealgranuloma: characteristics <strong>of</strong> <strong>the</strong> covering epi<strong>the</strong>lium. J LaryngolOtol 114:264–267218. MacGregor AR, Batsakis JG, El-Naggar AK (2003) My<strong>of</strong>ibroblastoma<strong>of</strong> <strong>the</strong> larynx: a study <strong>of</strong> two cases. <strong>Head</strong> <strong>Neck</strong>25:606–611219. MacMillan RH III, Fechner RE (1986) Pleomorphic adenoma<strong>of</strong> <strong>the</strong> larynx. Arch Pathol Lab Med 110:245–247220. Madrigal FM, Godoy LM, Daboin KP, Casiraghi O, GarciaAM, Lu MA (2002) Laryngeal osteosarcoma: a clinicopathologicanalysis <strong>of</strong> four cases <strong>and</strong> comparison with a carcinosarcoma.Ann Diagn Pathol 6:1–9221. Mahlstedt K, Uβmüller J, Donath K (2002) Malignant sialogenictumours <strong>of</strong> <strong>the</strong> larynx. J Laryngol Otol 116:119–122222. Maier H, Dietz A, Gewelke U, Heller WD, Weidauer H (1992)Tobacco <strong>and</strong> alcohol <strong>and</strong> <strong>the</strong> risk <strong>of</strong> head <strong>and</strong> neck cancer.Clin Invest 70:320–327223. Maier H, Tisch M (1997) Epidemiology <strong>of</strong> laryngeal cancer:results <strong>of</strong> <strong>the</strong> Heidelberg case-control study. Acta OtolaryngolSuppl 527:160–164224. Maier W, Lohle E, Welte V (1994) Pathogenetic <strong>and</strong> <strong>the</strong>rapeuticaspects <strong>of</strong> contact granuloma. Laryngorhinootologie73:488–491225. Mamelle G, Pampurik J, Luboinski B, Lancar R, Lusinchi A,Bosq J (1994) Lymph node prognostic factors in head <strong>and</strong>neck squamous cell carcinomas. Am J Surg 168:494–498226. Marcotullio D, Magliulo G, Pietrunti S, Suriano M (2002)Exudative laryngeal diseases <strong>of</strong> Reinke’s space: a clinicohistopathologicalframing. J Otolaryngol 31:376–380227. Marcotullio D, Paduano F, Magliulo G (1996) Laryngopyocele:an atypical case. Am J Otolaryngol 17:345–348228. Marion RB, Alperin JE, Maloney WH (1972) Gouty tophus <strong>of</strong><strong>the</strong> true vocal cord. Arch Otolaryngol 96:161–162229. Maronian NC, Azadeh H, Waugh P, Hillel A (2001) Association<strong>of</strong> laryngopharyngeal reflux disease <strong>and</strong> subglottic stenosis.Ann Otol Rhinol Laryngol 110:606–612230. Martin-Hirsch DP, Lannigan FJ, Irani B, Batman P (1992)Oncocytic papillary cystadenomatosis <strong>of</strong> <strong>the</strong> larynx. J LaryngolOtol 106:656–658231. Mayo-Smith MF, Spinale JW, Donskey CJ, Yukawa M, LiRH, Schiffman FJ (1995) Acute epiglottitis. An 18-year experiencein Rhode Isl<strong>and</strong>. Chest 108:1640–1647232. Mazur MT, Shultz JJ, Myers JL (1990) Granular cell tumor.Immunohistochemical analysis <strong>of</strong> 21 benign tumors <strong>and</strong> onemalignant tumor. Arch Pathol Lab Med 114:692–696


Larynx <strong>and</strong> Hypopharynx Chapter 7 231233. McAdam LP, O’Hanlan MA, Bluestone R, Pearson CM(1976) Relapsing polychondritis: prospective study <strong>of</strong> 23 patients<strong>and</strong> a review <strong>of</strong> <strong>the</strong> literature. Medicine 55:193–215234. McCluggage WC, Cameron CH, Arthure K, Toner PG (1997)Atypical carcinoid tumor <strong>of</strong> <strong>the</strong> larynx: an immunohistochemical,ultrastructural, <strong>and</strong> flow cytometric analysis. UltrastructPathol 21:431–438235. McGavran MH, Bauer WC, Ogura JH (1961) The incidence <strong>of</strong>cervical lymph node metastases from epidermoid carcinoma<strong>of</strong> <strong>the</strong> larynx <strong>and</strong> <strong>the</strong>ir relationship to certain characteristics<strong>of</strong> <strong>the</strong> primary tumor. A study based on <strong>the</strong> clinical <strong>and</strong> pathologicalfindings for 96 patients treated by primary en bloclaryngectomy <strong>and</strong> radical neck dissection. Cancer 14:55–66236. McGregor DK, Citron D, Shahab I (2003) Cryptococcal infection<strong>of</strong> <strong>the</strong> larynx simulating laryngeal carcinoma. SouthMed J 96:74–77237. McLaughlin RB, Spiegel JR, Selber J, Gotsdiner DB, Satal<strong>of</strong>fRT (1999) Laryngeal sarcoidosis presenting as an isolatedsubmucosal vocal fold mass. J Voice 13:240–245238. Michaels L, Hellquist HB (2001) Ear, nose <strong>and</strong> throat histopathology,2nd edn. Springer, London239. Michet CJ Jr, McKenna CH, Luthra HS, O’Fallon WM (1986)Relapsing polychondritis. Survival <strong>and</strong> predictive role <strong>of</strong> earlydisease manifestations. Ann Intern Med 104:74–78240. Mikaelian DO, Contrucci RB, Batsakis JG (1986) Epi<strong>the</strong>lial-myoepi<strong>the</strong>lialcarcinoma <strong>of</strong> <strong>the</strong> subglottic region: a casepresentation <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Otolaryngol <strong>Head</strong><strong>Neck</strong> Surg 95:104–106241. Miller FR, Wanamaker JR, Hicks DM, Tucker HM (1994)Cricoarytenoid arthritis <strong>and</strong> ankylosing spondylitis. ArchOtolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 120:214–216242. Mills SE (2002) Neuroendocrine neoplasms <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck with emphasis on neuroendocrine carcinomas. ModPathol 15:264–278243. Milroy CM, Rode J, Moss E (1991) Laryngeal paragangliomas<strong>and</strong> neuroendocrine carcinomas. Histopathology 18:201–209244. Mineta H, Miura K, Takebayashi S, Araki K, Ueda Y, HaradaH, Misawa K (2001) Immunohistochemical analysis <strong>of</strong> smallcell carcinoma <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck: a report <strong>of</strong> four patients<strong>and</strong> a review <strong>of</strong> sixteen patients in <strong>the</strong> literature with ectopichormone production. Ann Otol Rhinol Laryngol 110:76–82245. Mittal KR, True LD (1988) Origin <strong>of</strong> granules in granularcell tumor. Intracellular myelin formation with autodigestion.Arch Pathol Lab Med 112:302–303246. Mochloulis G, Irving RM, Grant HR, Miller RF (1996) LaryngealKaposi’s sarcoma in patients with AIDS. J LaryngolOtol 110:1034–1037247. Mohsenifar Z, Tashkin DP, Carson SA, Bellamy PE (1982)Pulmonary function in patients with relapsing polychondritis.Chest 81:711–717248. Moisa II, Silver CE (1991) Treatment <strong>of</strong> neuroendocrineneoplasms <strong>of</strong> <strong>the</strong> larynx. ORL J Otorhinolarngol Rel Spec53:259–264249. Mok JSW, Pak MW, Chan KF, Chow J, van Hasselt CA (2001)Unusual T- <strong>and</strong> T/NK- cell non-Hodgkin’s lymphoma <strong>of</strong> <strong>the</strong>larynx: a diagnostic challenge for clinicians <strong>and</strong> pathologists.<strong>Head</strong> <strong>Neck</strong>. 23:625–628250. Molloy AR, McMahon JN (1988) Rapid progression <strong>of</strong> trachealstenosis associated with tracheopathia osteo-chondroplastica.Intensive Care Med 15:60–62251. Monday LA, Cornut G, Bouchayer M, Roch JB (1983) Epidermoidcysts <strong>of</strong> <strong>the</strong> vocal cords. Ann Otol Rhinol Laryngol92:124–127252. Moore KL, Persaud TVN (1998) The developing human, 6<strong>the</strong>dn. Saunders, Philadelphia, pp 215–256253. Muir C, Weil<strong>and</strong> L (1995) Upper aerodigestive tract cancers.Cancer 75:147–153254. Muscat JE, Wynder EL (1992) Tobacco, alcohol, asbestos,<strong>and</strong> occupational risk factors for laryngeal cancer. Cancer69:2244–2251255. Myssiorek D, Halaas Y, Silver C (2001) Laryngeal <strong>and</strong> sinonasalparagangliomas. Otolaryngol Clin North Am 34:971–982256. Myssiorek D, Patel M, Wasserman P, R<strong>of</strong>eim O (1998) Osteosarcoma<strong>of</strong> <strong>the</strong> larynx. Ann Otol Rhinol Laryngol 107:70–74257. Myssiorek D, Persky M (1989) Laser endoscopic treatment<strong>of</strong> laryngoceles <strong>and</strong> laryngeal cysts. Otolaryngol <strong>Head</strong> <strong>Neck</strong>Surg 100:538–541258. Nakajima H (1993) Tuberculosis: a global emergency. WorldHealth Forum 14:438259. Nakayama M, Br<strong>and</strong>enburg JH, Hafez GR (1993) Dedifferentiatedchondrosarcoma <strong>of</strong> <strong>the</strong> larynx with regional <strong>and</strong> distantmetastases. Ann Otol Rhinol Laryngol 102:785–791260. N<strong>and</strong>apalan V, Rol<strong>and</strong> NJ, Helliwell TR, Williams EM, HamiltonJ, Jones AS (1998) Mucosal melanoma <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck. Clin Otolaryngol 23:107–116261. Narozny W, Stankiewicz C, Przewozny T, Bakowska A,Czuszynska Z (2001) A case <strong>of</strong> multisymptomatic relapsingpolychondritis in a 22-year-old woman. Acta OtorhinolaryngolBelg 55:227–233262. Nayak DR, Balakrishnan R, Rao RV, Hazarika P (2001) Clearcell carcinoma <strong>of</strong> <strong>the</strong> larynx. Int J Pediatr Otorhinolaryngol57:149–153263. Neel HB III, McDonald TJ (1982) Laryngeal sarcoidosis: report<strong>of</strong> 13 patients. Ann Otol Rhinol Laryngol 91:359–362264. Netterville JL, Coleman JR Jr, Chang S, Rainey CL, ReinischL, Oss<strong>of</strong>f RH (1998) Lateral laryngotomy for <strong>the</strong> removal <strong>of</strong>Teflon granuloma. Ann Otol Rhinol Laryngol 107:735–744265. Newman BH, Taxy JB, Laker HI (1984) Laryngeal cystsin adults: a clinicopathologic study <strong>of</strong> 20 cases. Am J ClinPathol 81:715–720266. Nguyen HC, Urquhart AC (1997) Zenker’s diverticulum. Laryngoscope107:1436–1440267. Nicolai P, Puxeddu R, Cappiello J, Peretti G, Battochio S,Facchetti F, Antonelli AR (1996) Metastatic neoplasms to <strong>the</strong>larynx: report <strong>of</strong> three cases. Laryngoscope 106:851–855268. Nishiike S, Irifune M, Doi K, Sawada T, Kubo T (2002) Laryngealtuberculosis: a report <strong>of</strong> 15 cases. Ann Otol RhinolLaryngol 111:916–918269. Nosfinger YC, Mirza N, Rowan PT, Lanza D, Weinstein G(1997) <strong>Head</strong> <strong>and</strong> neck manifestations <strong>of</strong> plasma cell neoplasms.Laryngoscope 107:741–746270. Nowak-Sadzikowska J, Weiss M (1998) Extramedullary plasmacytoma<strong>of</strong> <strong>the</strong> larynx. Analysis <strong>of</strong> 5 cases. Eur J Cancer34:1468271. Nunez DA, Astley SM, Lewis FA, Wells M (1994) Humanpapilloma viruses: a study <strong>of</strong> <strong>the</strong>ir prevalence in <strong>the</strong> normallarynx. J Laryngol Otol 108:319–320272. Ogawa Y, Nishiyama N, Hagiwara A, Ami T, Fujita H, YoshidaT, Suzuki M (2002) A case <strong>of</strong> laryngeal aspergillosis followingradiation <strong>the</strong>rapy. Auris Nasus Larynx 29:73–76273. O’Halloran LR, Lusk RP (1994) Amyloidosis <strong>of</strong> <strong>the</strong> larynx ina child. Ann Otol Rhinol Laryngol 103:590–594274. Ohlms LA, McGill T, Healy GB (1994) Malignant laryngealtumors in children: a 15-year experience with four patients.Ann Otol Rhinol Laryngol 103:686–692275. Omeroglu A, Weisenberg E, Baim HM, Rhone DP (2001)Pathologic quiz case. Supraglottic granulomas in a youngCentral American Man. Arch Pathol Lab Med 125:157–158276. Oreggia F, De Stefani E, B<strong>of</strong>fetta P, Brennan P, Deneo-PellegriniH, Ronco AL (2001) Meat, fat <strong>and</strong> risk <strong>of</strong> laryngealcancer: a case-control study in Uruguay. Oral Oncol 37:141–145277. Orl<strong>and</strong>i A, Fratoni S, Hermann I, Spagnoli LG (2003) Symptomaticlaryngeal nodular chondrometaplasia: a clinicopathologicalstudy. J Clin Pathol 56:976–977278. Osammor JY, Bulman CH, Blewitt RW (1990) Intralaryngotrachealthyroid. J Laryngol Otol 104:733–736279. Paaske PB, Tang E (1985) Tracheopathia osteoplastica in <strong>the</strong>larynx. J Laryngol Otol 99:305–310280. Pabuççuoğlu U, Tuncer C, Sengiz S (2002) Histopathology <strong>of</strong>c<strong>and</strong>idal hyperplastic lesions <strong>of</strong> <strong>the</strong> larynx. Pathol Res Pract198:675–678281. Park SS, Streitz JM Jr, Rebeiz EE, Shapshay SM (1995) Idiopathicsubglottic stenosis. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg121:894–897


232 N. Gale · A. Cardesa · N. Zidar7282. Patterson HC, Dickerson GR, Pilch BZ, Bentkover SH (1981)Hamartoma <strong>of</strong> <strong>the</strong> hypopharynx. Arch Otolaryngol 107:767–772283. Peeters A, Schmelzer B, Beernaert A, Fannes H (2001) Oncocyticlaryngeal cysts: a case report <strong>and</strong> literature review. ActaOtorhinolaryngol Belg 55:71–75284. Pelliteri PK, Ferlito A, Bradley PJ, Shaha AR, Rinaldo A(2003) Management <strong>of</strong> sarcomas <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck inadults. Oral Oncol 39:2–12285. Pelucchi C, Talamini R, Levi F, Bosetti C, La Vecchia C, NegriE, Parpinel M, Pesce C, Tobia-Gallelli F, Toncini C (1984)APUD cells <strong>of</strong> <strong>the</strong> larynx. Acta Otolaryngol 98:158–162286. Pelucchi S, Amoroso C, Gr<strong>and</strong>i E, Carinci F, Pastore A(2002) Granular cell tumour <strong>of</strong> <strong>the</strong> larynx: literature review<strong>and</strong> case report. J Otolaryngol 31:234–235287. Pennings RJ, van den Hoogen FJ, Marres HA (2001) Giantlaryngoceles: a cause <strong>of</strong> upper airway obstruction. Eur ArchOtorhinolaryngol 258:137–140288. Pesce C, Tobia-Gallelli F, Toncini C (1984) APUD cells <strong>of</strong> <strong>the</strong>larynx. Acta Otolaryngol 98:158–162289. Peterson KL, Fu YS, Calcaterra T (1997) Subglottic paraganglioma.<strong>Head</strong> <strong>Neck</strong> 19:54–56290. Phipps CD, Gibson WS, Wood WE (1997) Infantile subglottichemangioma: a review <strong>and</strong> presentation <strong>of</strong> two cases <strong>of</strong>surgical excision. Int J Pediatr Otorhinolaryngol 41:71–79291. Pilch BZ (2001) Larynx <strong>and</strong> hypopharynx. In: Pilch BZ (ed)<strong>Head</strong> <strong>and</strong> neck surgical pathology. Lippincott Williams &Wilkins, Philadelphia, pp 230–283292. Pinsolle J, Pinsolle V, Majoufre C, Duroux S, Demeaux H, SiberchicotF (1997) Prognostic value <strong>of</strong> histologic findings inneck dissections for squamous cell carcinoma. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 123:145–148293. Pompe-Kirn V (2002) Epidemiological features <strong>of</strong> laryngealcancer in Slovenia. Zdrav Vestn 71:59–63294. Popper HH, Winter E, H<strong>of</strong>ler G (1994) DNA <strong>of</strong> Mycobacteriumtuberculosis in formalin-fixed, paraffin-embedded tissuein tuberculosis <strong>and</strong> sarcoidosis detected by polymerase chainreaction. Am J Clin Pathol 101:738–741295. Pounder DJ, Pieterse AS (1982) Tracheopathia osteoplastica:a study <strong>of</strong> <strong>the</strong> minimal lesion. J Pathol 138:235–239296. Prescher A, Schick B, Stütz A, Brors D (2002) Laryngeal prostaticcancer metastases: an underestimated route <strong>of</strong> metastases?Laryngoscope 112:1467–1473297. Preti G, Palonta F, Vione N, Rosso P, Cavalot AL (2003) Leiomyosarcoma<strong>of</strong> <strong>the</strong> larynx. Tumori 89:321–323298. Pribitkin E, Friedman O, O’Hara B, Cunnane MF, Levi D,Rosen M, Keane WM, Satal<strong>of</strong>f RT (2003) Amyloidosis <strong>of</strong> <strong>the</strong>upper aerodigestive tract. Laryngoscope 113:2095–2101299. Rafferty MA, Fenton JE, Jones AS (2001) The history, aetiology<strong>and</strong> epidemiology <strong>of</strong> laryngeal carcinoma. Clin Otolaryngol26:442–446300. Raitiola H, Puk<strong>and</strong>er J (2000) Symptoms <strong>of</strong> laryngeal carcinoma<strong>and</strong> <strong>the</strong>ir prognostic significance. Acta Oncol 39:213–216301. Raitiola H, Puk<strong>and</strong>er J, Laippala P (1999) Glottic <strong>and</strong> supraglotticlaryngeal carcinoma: differences in epidemiology,clinical characteristics <strong>and</strong> prognosis. Acta Otolaryngol119:847–851302. Rajah V, Essa A (1993) Histoplasmosis <strong>of</strong> <strong>the</strong> oral cavity, oropharynx<strong>and</strong> larynx. J Laryngol Otol 107:58–61303. Rakover Y, Bennett M, David R, Rosen G (2000) Isolated extramedullaryplasmacytoma <strong>of</strong> <strong>the</strong> vocal fold. J LaryngolOtol 114:540–542304. Ramesar K, Albizzati C (1988) Laryngeal cysts: clinical relevance<strong>of</strong> a modified working classification. J Laryngol Otol102:923–925305. R<strong>and</strong>olph J, Grunt JA, Vawter GF (1963) The medical <strong>and</strong> surgicalaspects <strong>of</strong> intratracheal goiter. N Engl J Med 268:457–461306. Rassaei N, Frye DA, Harter KW, Troost TR, Ozdemirli M(2003) Acinic cell carcinoma <strong>of</strong> <strong>the</strong> glottis: case report. Am JOtolaryngol 24:258–260307. Reder PA, Neel HB III (1993) Blastomycosis in otolaryngology:review <strong>of</strong> a large series. Laryngoscope 103:53–58308. Remacle M, Degols JC, Delos M (1996) Exudative lesions <strong>of</strong>Reinke’s space. An anatomopathological correlation. ActaOtorhinolaryngol Belg 50:253–264309. Remenar E, Elo J, Frint T (1984) The morphological basis fordevelopment <strong>of</strong> Reinke’s oedema. Acta Otolaryngol 97:169–176310. Richardson BE, Morrison VA, Gapany M (1996) Invasive aspergillosis<strong>of</strong> <strong>the</strong> larynx: case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 114:471–473311. Richter B, Fradis M, Kohler G, Ridder GJ (2001) Epiglottictuberculosis: differential diagnosis <strong>and</strong> treatment. Case report<strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Ann Otol Rhinol Laryngol110:197–201312. Riding K (1992) Subglottic hemangioma: a practical approach.J Otolaryngol 21:419–421313. Rihkanen H, Peltomaa J, Syrjänen S (1994) Prevalence <strong>of</strong> humanpapillomavirus DNA in vocal cords without laryngealpapillomas. Acta Otolaryngol 114:348–351314. Rinaldo A, Mannara GM, Fisher C, Ferlito A (1998) Hamartoma<strong>of</strong> <strong>the</strong> larynx: a critical review <strong>of</strong> <strong>the</strong> literature. AnnOtol Rhinol Laryngol 107:264–267315. Rizzo PB, Da Mosto MC, Clari M, Scotton PG, Vaglia A,Marchiori C (2003) Laryngeal tuberculosis: an <strong>of</strong>ten forgottendiagnosis. Int J Infect Dis 7:129–131316. Salvesen R, Brautaset NJ (1996) Oculopharyngeal musculardystrophy in Norway. Survey <strong>of</strong> a large Norwegian family.Acta Neurol Sc<strong>and</strong> 93:281–285317. Sant’Anna GD, Mauri M, Arrarte JL, Camargo H Jr (1999)Laryngeal manifestations <strong>of</strong> paracoccidioidomycosis (SouthAmerican blastomycosis). Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg125:1375–1378318. Santoro R, Turelli M, Polli G (2000) Primary carcinoma <strong>of</strong><strong>the</strong> subglottic larynx. Eur Arch Otorhinolaryngol 257:548–551319. Satal<strong>of</strong>f RT, Wilborn A, Prestipino A, Hawkshaw M, HeuerRJ, Cohn J (1993) Histoplasmosis <strong>of</strong> <strong>the</strong> larynx. Am J Otolaryngol14:199–205320. Sato K, Hirano M, Nakashima T (1999) Electron microscopic<strong>and</strong> immunohistochemical investigation <strong>of</strong> Reinke’s edema.Ann Otol Rhinol Laryngol 108:1068–1072321. Sato K, Kurita S, Hirano M (1993) Location <strong>of</strong> <strong>the</strong> preepiglotticspace <strong>and</strong> its relationship to <strong>the</strong> paraglottic space. AnnOtol Rhinol Laryngol 102:930–934322. Sato K, Umeno T, Hirano M, Nakashima T (2002) Cricoidarea <strong>of</strong> <strong>the</strong> larynx: its physiological <strong>and</strong> pathological significance.Acta Otolaryngol 122:882–886323. Sawatsubashi M, Tuda K, Tokunaga O, Shin T (1997) Pleomorphicadenoma <strong>of</strong> <strong>the</strong> larynx: a case <strong>and</strong> review <strong>of</strong> <strong>the</strong> literaturein Japan. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 117:415–417324. Saydam L, Koybasi S, Kutluay L (2003) Laryngeal chondromapresenting as an external neck mass. Eur Arch Otorhinolaryngol260:239–241325. Schiff NF, Annino DJ, Woo P, Shapshay SM (1997) Kaposi’ssarcoma <strong>of</strong> <strong>the</strong> larynx. Ann Otol Rhinol Laryngol 106:563–567326. Schmidt U, Metz KA, Schrader M, Leder LD (1994) Well-differentiated(oncocytoid) neuroendocrine carcinoma <strong>of</strong> <strong>the</strong>larynx with multiple skin metastases – a brief report. J LaryngolOtol 108:272–274327. See AC, Patel SG, Montgomery PQ, Rhys Evans PH, Fisher C(1998) Intralaryngotracheal thyroid-ectopic thyroid or invasivecarcinoma? J Laryngol Otol 112:673–676328. Seikaly H, Cuyler JP (1994) Infantile subglottic hemangioma.J Otolaryngol 23:135–137329. Seitz HK, Simanowski UA, Kommerell B (1988) Alcohol <strong>and</strong>cancer. Z Gastroenterol Suppl 3:106–119330. Shapiro AM, Rimell FL, Kenna MA (1995) Pathologic quizcase 1. Granular cell tumor (GCT) <strong>of</strong> larynx. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 121:1058; 1060–1061331. Shapiro J, Eavey RD, Baker AS (1988) Adult supraglottitis. Aprospective analysis. JAMA 259:563–567332. Sheen TS, Wu CT, Heieh T, Hsu MM (1997) Postirradiationlaryngeal osteosarcoma: case report <strong>and</strong> literature review.<strong>Head</strong> <strong>Neck</strong> 19:57–62


Larynx <strong>and</strong> Hypopharynx Chapter 7 233333. Shek AW, Wu PC, Samman N (1996) Inflammatory pseudotumour<strong>of</strong> <strong>the</strong> mouth <strong>and</strong> maxilla. J Clin Pathol 49:164–167334. Shikhani AH, Jones MM, Marsh BR, Holliday MJ (1986) Infantilesubglottic hemangiomas. An update. Ann Otol RhinolLaryngol 95:336–347335. Shonai T, Hareyama M, Sakata K, Oouchi A, Nagakura H,Koito K, Morita K, Satoh M, Asakura K, Kataura A, HinodaY (1998) Mucoepidermoid carcinoma <strong>of</strong> <strong>the</strong> larynx: a casewhich responded completely to radio<strong>the</strong>rapy <strong>and</strong> a review <strong>of</strong><strong>the</strong> literature. Jpn J Clin Oncol 28:339–342336. Shpitzer T, Noyek AM, Witterick I, Kassel T, Ichise M,Gullane P, Neligan P, Freeman J (1997) Noncutaneous cavernoushemangiomas <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Am J Otolaryngol18:367–374337. Sie KC, McGill T, Healy GB (1994) Subglottic hemangioma:ten years’ experience with <strong>the</strong> carbon dioxide laser. Ann OtolRhinol Laryngol 103:167–172338. Smets G, Warson F, Dehou MF, Storme G, Sacre R, Van BelleS, Somers G, Gepts W, Kloppel G (1990) Metastasizing neuroendocrinecarcinoma <strong>of</strong> <strong>the</strong> larynx with calcitonin <strong>and</strong> somatostatinsecretion <strong>and</strong> CEA production, resembling medullarythyroid carcinoma. Virchows Arch 416:539–543339. Sneller MC (1995) Wegener’s granulomatosis. JAMA273:1288–1291340. Sobin LH, Wittekind CH (2002) TNM classification <strong>of</strong> malignanttumors, 6th edn, Wiley, New York341. Soga J, Osaka M, Yakuwa Y (2002) Laryngeal endocrinomas(carcinoids <strong>and</strong> relevant neoplasms): analysis <strong>of</strong> 278 reportedcases. J Exp Cancer Res 21:5–13342. Soni NK (1992) Leprosy <strong>of</strong> <strong>the</strong> larynx. J Laryngol Otol106:518–520343. Sorensen WT, Moller-Andersen K, Behrendt N (1998) Rheumatoidnodules <strong>of</strong> <strong>the</strong> larynx. J Laryngol Otol 112:573–574344. Souglu Y, Erdamar B, Katircioglu OS, Karatay MC, Sunay T(2002) Extracapsular spread in ipsilateral neck <strong>and</strong> contralateralneck metastases in laryngeal cancer. Ann Otol RhinolLaryngol 111:447–454345. Soylu L, Kiroglu F, Ersoz C, Ozcan C, Aydogan B (1993) Intralaryngotrachealthyroid. Am J Otolaryngol 14:145–147346. Spector JG, Sessions DG, Emami B, Simpson J, Haughey B,Harvey J, Fredrickson JM (1995) Squamous cell carcinoma <strong>of</strong><strong>the</strong> pyriform sinuses: a nonr<strong>and</strong>omized study comparison <strong>of</strong><strong>the</strong>rapeutic modalities <strong>and</strong> long-term results. Laryngoscope105:397–406347. Spiro RH (1986) Salivary neoplasms: overview <strong>of</strong> a 35-yearexperience with 2807 patients. <strong>Head</strong> <strong>Neck</strong> Surg 8:177–184348. Spiro RH (1997) Distant metastasis in adenoid cystic carcinoma<strong>of</strong> <strong>the</strong> salivary origin. Am J Surg 174:495–498349. Spraggs PD, Tostevin PM, Howard DJ (1997) Management <strong>of</strong>laryngotracheobronchial sequelae <strong>and</strong> complications <strong>of</strong> relapsingpolychondritis. Laryngoscope 107:936–941350. Stell PM, Tobin KE (1975) The behaviour <strong>of</strong> cancer affecting<strong>the</strong> subglottic space. Can J Otolaryngol 4:612–617351. Strickler JG, Audeh MW, Copenhaver CM, Warnke RA(1988) Immunophenotypic differences between plasmacytoma,multiple myeloma <strong>and</strong> immunoblastic lymphoma. Cancer61:1782–1786352. Strong MS, Vaughan CW (1971) Vocal cord nodules <strong>and</strong> polyps– <strong>the</strong> role <strong>of</strong> surgical treatment. Laryngoscope 81:911–923353. Szpunar J, Glowacki J, Laskowski A, Miszke A (1971) Fibrinouslaryngotracheobronchitis in children. Arch Otolaryngol93:173–178354. Szwarc BJ, Kashima HK (1997) Endoscopic management <strong>of</strong> acombined laryngocele. Ann Otol Rhinol Laryngol 106:556–559355. Šmid L, Lavrenčak B, Žargi M (1992) Laryngo-tracheo-bronchopathiachondro-osteoplastica. J Laryngol Otol 106:845–848356. Tajima K, Yamakawa M, Katagiri T, Sasaki H (1997) Immunohistochemicaldetection <strong>of</strong> bone morphogenetic protein-2<strong>and</strong> transforming growth factor beta-1 in tracheopathia osteochondroplastica.Virchows Arch 431:359–363357. Thabet MH, Kotob H (2001) Lateral saccular cysts <strong>of</strong> <strong>the</strong> larynx.Aetiology, diagnosis <strong>and</strong> management. J Laryngol Otol115:293–297358. Thome R, Thome DC, de la Cortina RA (2001) Long-termfollow-up <strong>of</strong> cartilaginous tumors <strong>of</strong> <strong>the</strong> larynx. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 124:634–640359. Thompson LD, Derringer GA, Wenig BM (2000) Amyloidosis<strong>of</strong> <strong>the</strong> larynx: a clinicopathologic study <strong>of</strong> 11 cases. ModPathol 13:528–535360. Thompson LDR, Gannon FH (2002) Chondrosarcoma <strong>of</strong> <strong>the</strong>larynx. A clinicopathologic study <strong>of</strong> 111 cases with review <strong>of</strong><strong>the</strong> literature. Am J Surg Pathol 26:836–851361. Thoren L (1947) Intratracheal goitre. Acta Chir Sc<strong>and</strong>95:495–512362. Tisch M, Lampl L, Groh A, Maier H (2002) Angioneuroticedemas <strong>of</strong> <strong>the</strong> upper aerodigestive tract after ACE-inhibitortreatment. Eur Arch Otorhinolaryngol 259:419–421363. Toohill RJ (1975) The psychosomatic aspects <strong>of</strong> children withvocal nodules. Arch Otolaryngol 101:591–595364. Trentham DE, Le CH (1998) Relapsing polychondritis. AnnIntern Med 129:114–122365. Tsikoudas A, Coatesworth AP, Martin-Hirsch DP (2002) Laryngealgout. J Laryngol Otol 116:140–142366. Tsunoda K, Soda Y (1996) Hoarseness as <strong>the</strong> initial manifestation<strong>of</strong> systemic lupus ery<strong>the</strong>matosus. J Laryngol Otol110:478–479367. Tuyns AJ, Estève J, Raymond L, Berrino F, Benhamou E,Blanchet F, B<strong>of</strong>fetta P, Crosignani P, del Moral A, LehmannW, Merletti F, Péquignot G, Riboli E, Sancho-Garnier H, TerraciniB, Zubiri A, Zubiri L (1988) Cancer <strong>of</strong> <strong>the</strong> larynx/hypopharynx,tobacco <strong>and</strong> alcohol: IARC international casecontrolstudy in Turin <strong>and</strong> Varese (Italy), Zaragoza <strong>and</strong> Navarra(Spain), Geneva (Switzerl<strong>and</strong>) <strong>and</strong> Calvados (France).Int J Cancer 41:483–491368. Uzcudun EA, Fern<strong>and</strong>ez PB, Sanchez JJ, Gr<strong>and</strong>e AG, RetolazaIR, Baron MG, Bouzas JG (2001) Clinical features <strong>of</strong> pharyngealcancer: a retrospective study <strong>of</strong> 258 consecutive patients.J Laryngol Otol 115:112–118369. Valdez TA, Shapshay SM (2002) Idiopathic subglottic stenosisrevisited. Ann Otol Rhinol Laryngol 111:690–695370. Varvares MA, Montgomery WW, Hillman RE (1995) Teflongranuloma <strong>of</strong> <strong>the</strong> larynx: etiology, pathophysiology, <strong>and</strong>management. Ann Otol Rhinol Laryngol 104:511–515371. Victoria LV, H<strong>of</strong>fman HT, Robinson RA (1998) Granular celltumour <strong>of</strong> <strong>the</strong> larynx. J Laryngol Otol 112:373–376372. Waggoner LG (1958) Intralaryngeal intratracheal thyroid.Ann Otol Rhinol Laryngol 67:61–71373. Walker GK, Fechner RE, Johns ME, Teja K (1982) Necrotizingsialometaplasia <strong>of</strong> <strong>the</strong> larynx secondary to a<strong>the</strong>romatousembolization. Am J Clin Pathol 77:221–223374. Wani MK, Woodson GE (1999) Laryngeal contact granuloma.Laryngoscope 109:1589–1593375. Wasserman PG, Savargaonkar P (2001) Paragangliomas:classification, pathology, <strong>and</strong> differential diagnosis. OtolaryngolClin North Am 34:845–862376. Waterhouse JAH (1993) Epidemiology. In: Ferlito A (ed)Neoplasms <strong>of</strong> <strong>the</strong> larynx. Churchill Livingstone, Edinburgh,pp 49–64377. Weber AL, Shortsleeve M, Goodman M, Montgomery W,Grillo HC (1978) Cartilaginous tumors <strong>of</strong> <strong>the</strong> larynx <strong>and</strong> trachea.Radiol Clin North Am 16:261–267378. Welsh J, Westra WH, Eisele D, Hogan R, Lee DJ (1998) Solitaryplasmacytoma <strong>of</strong> <strong>the</strong> epiglottis: a case report <strong>and</strong> review<strong>of</strong> <strong>the</strong> literature. J Laryngol Otol 112:174–176379. Wenig BM (1995) Laryngeal mucosal malignant melanoma.A clinicopathologic, immunohistochemical, <strong>and</strong> ultrastructuralstudy <strong>of</strong> four patients <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Cancer75:1568–1577380. Wenig BM (1995) Necrotizing sialometaplasia <strong>of</strong> <strong>the</strong> larynx.A report <strong>of</strong> two cases <strong>and</strong> a review <strong>of</strong> <strong>the</strong> literature. Am J ClinPathol 103:609–613381. Wenig BM, Abbondanzo SL, Childers EL, Kapadia SB, HeffnerDR (1993) Extranodal sinus histiocytosis with massivelymphadenopathy (Rosai-Dorfman disease) <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck. Hum Pathol 24:483–492


234 N. Gale · A. Cardesa · N. Zidar7382. Wenig BM, Devaney K, Bisceglia M (1995) Inflammatorymy<strong>of</strong>ibroblastic tumor <strong>of</strong> <strong>the</strong> larynx. A clinicopathologicstudy <strong>of</strong> eight cases simulating a malignant spindle cell neoplasm.Cancer 76:2217–2229383. Wenig BM, Devaney K, Wenig BL (1995) Pseudoneoplasticlesions <strong>of</strong> <strong>the</strong> oropharynx <strong>and</strong> larynx simulating cancer.Pathol Annu 30:143–187384. Wenig BM, Heffner DK (1990) Contact ulcers <strong>of</strong> <strong>the</strong> larynx.A reacquaintance with <strong>the</strong> pathology <strong>of</strong> an <strong>of</strong>ten underdiagnosedentity. Arch Pathol Lab Med 114:825–828385. Wenig BM, Heffner DK, Oertel YC, Johnson FB (1990) Teflonomas<strong>of</strong> <strong>the</strong> larynx <strong>and</strong> neck. Hum Pathol 21:617–623386. Werkhaven J, Oss<strong>of</strong>f RH (1991) Surgery for benign lesions <strong>of</strong><strong>the</strong> glottis. Otolaryngol Clin North Am 24:1179–1199387. Westerberg BD, Durham JS, Berean KW (1995) Multiple oncocyticlaryngeal cysts presenting as acute airway obstruction.J Otolaryngol 24:319–321388. Woodruff JM, Senie RT (1991) Atypical carcinoid tumor <strong>of</strong><strong>the</strong> larynx. A critical review <strong>of</strong> <strong>the</strong> literature. ORL 53:194–209389. World Health Organization Classification <strong>of</strong> Tumours (2005)<strong>Pathology</strong> <strong>and</strong> genetics <strong>of</strong> tumours <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck.IARC, Lyon390. Yencha MW, Linfesty R, Blackmon A (2000) Laryngeal tuberculosis.Am J Otolaryngol 21:122–126391. Yuan YG, Han DM, Yang BQ, Yu ZK (1999) Laryngeal carcinoidtumours: Report <strong>of</strong> three cases. J Otolaryngol 28:54–56392. Yuen APW, Wei WI, Ho WK, Hui Y (1996) Risk factors <strong>of</strong>tracheostomal recurrence after laryngectomy for laryngealcarcinoma. Am J Surg 172:263–266393. Zatonski W, Becher H, Lissowska J, Wahrendorf J (1991) Tobacco,alcohol, <strong>and</strong> diet in <strong>the</strong> etiology <strong>of</strong> laryngeal cancer: apopulation-based case-control study. Cancer Causes Control2:3–10394. Zbaren P, Greiner R, Kengelbacher M (1996) Stomal recurrenceafter laryngectomy: an analysis <strong>of</strong> risk factors. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 114:569–575395. Zeitels SM, Hillman RE, Bunting GW, Vaughn T (1997) Reinke’sedema: phonatory mechanisms <strong>and</strong> managementstrategies. Ann Otol Rhinol Laryngol 106:533–543396. Zheng W, Blot WJ, Shu X, Gao YT, Ji BT, Ziegler RG, FraumeniJr (1992) Diet <strong>and</strong> o<strong>the</strong>r risk factors for laryngeal cancer inShanghai, China. Am J Epidemiol 136:178–191397. Zidar N, Volavšek M, Trček C, Kern I, Gale N (2000) Wegener’sgranulomatosis in <strong>the</strong> upper respiratory tract. Wien KlinWochenschr 112:676–679


Chapter 8Ear<strong>and</strong> Temporal BoneL. Michaels8Contents8.1 Summary <strong>of</strong> Embryology,Anatomy <strong>and</strong> Histology . . . . . . . . . . . . . . . . . 2368.1.1 Embryology . . . . . . . . . . . . . . . . . . . . . . . . 2368.1.2 Anatomy . . . . . . . . . . . . . . . . . . . . . . . . . . 2368.1.3 Histology . . . . . . . . . . . . . . . . . . . . . . . . . . 2378.2 External Ear <strong>and</strong> Auditory Canal . . . . . . . . . . . . 2378.2.1 Inflammatory <strong>and</strong> Metabolic Lesions . . . . . . . . . 2378.2.1.1 Diffuse External Otitis . . . . . . . . . . . . . . . . . . 2378.2.1.2 Perichondritis . . . . . . . . . . . . . . . . . . . . . . . 2378.2.1.3 Malignant Otitis Externa . . . . . . . . . . . . . . . . . 2378.2.1.4 Relapsing Polychondritis . . . . . . . . . . . . . . . . . 2388.2.1.5 Gout . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2388.2.1.6 Ochronosis . . . . . . . . . . . . . . . . . . . . . . . . . 2388.2.2 Pseudocystic <strong>and</strong> Cystic Lesions . . . . . . . . . . . . 2388.2.2.1 Idiopathic Pseudocystic Chondromalacia . . . . . . . 2388.2.2.2 First Branchial Cleft Cyst . . . . . . . . . . . . . . . . . 2388.2.3 Tumour-Like Lesions . . . . . . . . . . . . . . . . . . . 2398.2.3.1 Chondrodermatitis Nodularis Helicis . . . . . . . . . 2398.2.3.2 Keratosis Obturans <strong>and</strong> Cholesteatoma<strong>of</strong> External Canal . . . . . . . . . . . . . . . . . . . . . 2398.2.3.3 Keratin Granuloma . . . . . . . . . . . . . . . . . . . . 2398.2.3.4 Angiolymphoid Hyperplasiawith Eosinophilia <strong>and</strong> Kimura’s Disease . . . . . . . . 2398.2.3.5 Accessory Tragus . . . . . . . . . . . . . . . . . . . . . 2408.2.3.6 Keloid . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2408.2.4 Benign Neoplasms . . . . . . . . . . . . . . . . . . . . . 2408.2.4.1 Adenoma <strong>of</strong> Ceruminal Gl<strong>and</strong>s . . . . . . . . . . . . . 2408.2.4.2 Pleomorphic Adenoma<strong>of</strong> Ceruminal Gl<strong>and</strong>s . . . . . . . . . . . . . . . . . . . 2418.2.4.3 Syringocystadenoma Papilliferum<strong>of</strong> Ceruminal Gl<strong>and</strong>s . . . . . . . . . . . . . . . . . . . 2418.2.4.4 Bony Lesions . . . . . . . . . . . . . . . . . . . . . . . . 2418.2.5 Malignant Neoplasms . . . . . . . . . . . . . . . . . . . 2428.2.5.1 Adenocarcinoma <strong>of</strong> Ceruminal Gl<strong>and</strong>s . . . . . . . . 2428.2.5.2 Adenoid Cystic Carcinoma<strong>of</strong> Ceruminal Gl<strong>and</strong>s . . . . . . . . . . . . . . . . . . . 2428.2.5.3 Basal Cell Carcinoma . . . . . . . . . . . . . . . . . . . 2428.2.5.4 Squamous Cell Carcinoma . . . . . . . . . . . . . . . . 2438.2.5.5 Melanotic Neoplasms . . . . . . . . . . . . . . . . . . . 2438.3 Middle Ear <strong>and</strong> Mastoid . . . . . . . . . . . . . . . . . 2448.3.1 Inflammatory Lesions . . . . . . . . . . . . . . . . . . 2448.3.1.1 Acute <strong>and</strong> Chronic Otitis Media . . . . . . . . . . . . 2448.3.1.2 Cholesteatoma . . . . . . . . . . . . . . . . . . . . . . . 2448.3.1.3 Unusual Inflammatory Lesions . . . . . . . . . . . . . 2478.3.2 Neoplasms <strong>and</strong> LesionsResembling Neoplasms . . . . . . . . . . . . . . . . . . 2478.3.2.1 Choristoma(Salivary Gl<strong>and</strong>, Glial <strong>and</strong> Sebaceous Types) . . . . . 2478.3.2.2 Adenoma . . . . . . . . . . . . . . . . . . . . . . . . . . 2478.3.2.3 Papillary Tumours . . . . . . . . . . . . . . . . . . . . . 2488.3.2.4 Jugulotympanic Paraganglioma . . . . . . . . . . . . . 2498.3.2.5 Squamous Carcinoma . . . . . . . . . . . . . . . . . . 2508.3.2.6 Meningioma . . . . . . . . . . . . . . . . . . . . . . . . 2518.3.2.7 Rhabdomyosarcoma . . . . . . . . . . . . . . . . . . . 2518.3.2.8 Metastatic Carcinoma . . . . . . . . . . . . . . . . . . 2528.4 Inner Ear . . . . . . . . . . . . . . . . . . . . . . . . . . 2528.4.1 Bony Labyrinth . . . . . . . . . . . . . . . . . . . . . . 2528.4.1.1 Otosclerosis . . . . . . . . . . . . . . . . . . . . . . . . 2528.4.1.2 Paget’s Disease . . . . . . . . . . . . . . . . . . . . . . . 2538.4.1.3 Osteogenesis Imperfecta . . . . . . . . . . . . . . . . . 2548.4.1.4 Osteopetrosis . . . . . . . . . . . . . . . . . . . . . . . . 2548.4.2 Membranous Labyrinth <strong>and</strong> Cranial Nerves . . . . . 2548.4.2.1 Viral, Bacterial <strong>and</strong> Mycotic Infections . . . . . . . . 2548.4.2.2 Lesions <strong>of</strong> <strong>the</strong> Vestibular System . . . . . . . . . . . . 2568.4.2.3 Tumours <strong>and</strong> Tumour-Like Lesions . . . . . . . . . . 2578.4.2.4 Presbyacusis . . . . . . . . . . . . . . . . . . . . . . . . 2608.4.2.5 Malformations . . . . . . . . . . . . . . . . . . . . . . . 260References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 260


236 L. Michaels88.1 Summary <strong>of</strong> Embryology,Anatomy <strong>and</strong> Histology8.1.1 EmbryologyThe ear is not a single organ, but two, being <strong>the</strong> peripheralreceptor site both for stimuli derived from soundwaves <strong>and</strong> for changes in posture. The structures subservingboth <strong>of</strong> those functions are developed from aninvagination <strong>of</strong> <strong>the</strong> ectoderm early in embryonic life– <strong>the</strong> otocyst – to produce <strong>the</strong> epi<strong>the</strong>lia <strong>of</strong> <strong>the</strong> membranouslabyrinth <strong>of</strong> <strong>the</strong> inner ear. Superimposed upon,<strong>and</strong> developing slightly later, <strong>the</strong> first <strong>and</strong> second branchialarch systems provide structures that augment <strong>the</strong>hearing function. The endodermal component <strong>of</strong> <strong>the</strong>first branchial system, <strong>the</strong> branchial pouch, gives riseto <strong>the</strong> Eustachian tube <strong>and</strong> middle ear epi<strong>the</strong>lia <strong>and</strong> <strong>the</strong>corresponding ectodermal outgrowth, <strong>the</strong> first branchialcleft, to <strong>the</strong> external ear epidermis. The connectivetissue part <strong>of</strong> <strong>the</strong> local branchial cranial (auditory vestibular)nerve outflow from <strong>the</strong> central nervous system,both its vestibular <strong>and</strong> cochlear branches, grows to linkup with <strong>the</strong> sensory epi<strong>the</strong>lia lining <strong>the</strong> otocyst-derivedcochlear <strong>and</strong> vestibular endolymph-containing cavities;<strong>the</strong>re is recent evidence that terminal ganglion cells, e.g.spiral ganglion cells, may also be otocyst-derived. Cartilaginous,bony <strong>and</strong> muscular configurations <strong>of</strong> <strong>the</strong> earare developed from <strong>the</strong> mesenchyme surrounding <strong>the</strong>seearly epi<strong>the</strong>lia. The seventh cranial (facial) nerve developsin close relation to <strong>the</strong> structures <strong>of</strong> <strong>the</strong> ear for much<strong>of</strong> its course.8.1.2 AnatomyThe anatomy <strong>of</strong> <strong>the</strong> ear may be considered by referenceto its functions in hearing <strong>and</strong> balance (Fig. 8.1).The pinna <strong>and</strong> external canal conduct sound wavesin <strong>the</strong> air to <strong>the</strong> tympanic membrane, which transmits<strong>the</strong>m by very delicate vibrations. The middle ear enhancesthis sound energy transmission by conveying vibrationsfrom <strong>the</strong> larger area <strong>of</strong> <strong>the</strong> tympanic membranethrough <strong>the</strong> ossicular chain (malleus, incus <strong>and</strong> stapes)to <strong>the</strong> much smaller area <strong>of</strong> <strong>the</strong> footplate <strong>of</strong> <strong>the</strong> stapes,which lies in <strong>the</strong> oval window <strong>of</strong> <strong>the</strong> vestibule in contactwith <strong>the</strong> perilymph. In this way vibrations representingsound are conducted to <strong>the</strong> fluids <strong>of</strong> <strong>the</strong> inner ear. Theair space <strong>of</strong> <strong>the</strong> middle ear cavity is magnified by <strong>the</strong>mastoid air cells, which are complex expansions into <strong>the</strong>mastoid bone. There is a connection <strong>of</strong> <strong>the</strong> middle earspace with <strong>the</strong> nasopharynx <strong>and</strong> so with <strong>the</strong> external airthrough <strong>the</strong> Eustachian tube, by which air pressure canbe adjusted.From <strong>the</strong> vestibular perilymph, vibrations derivedfrom sound waves pass directly via <strong>the</strong> scala vestibuliinto <strong>the</strong> spirally coiled perilymphatic spaces <strong>of</strong> <strong>the</strong> cochlea,where it forms an upper compartment ascendingfrom <strong>the</strong> vestibule <strong>and</strong> oval window. A lower compartment,<strong>the</strong> scala tympani, descends spirally to <strong>the</strong> roundwindow membrane, a connective tissue disc separating<strong>the</strong> perilymph compartment from <strong>the</strong> middle ear. Between<strong>the</strong> scalae vestibuli <strong>and</strong> tympani <strong>the</strong>re is an endolymph-containingcoiled middle compartment, <strong>the</strong>cochlear duct (scala media), which houses <strong>the</strong> senso-Fig. 8.1. Anatomical diagram <strong>of</strong> <strong>the</strong>ear. Reproduced from Michaels <strong>and</strong>Hellquist [68]Tympanicmembrane


Ear <strong>and</strong> Temporal Bone Chapter 8 237ry organ <strong>of</strong> sound reception, <strong>the</strong> organ <strong>of</strong> Corti. Waves<strong>of</strong> vibration are conveyed from <strong>the</strong> perilymph to <strong>the</strong>walls <strong>of</strong> <strong>the</strong> scala media, from which, through <strong>the</strong> endolymph,<strong>the</strong>y affect <strong>the</strong> sensory cells <strong>of</strong> <strong>the</strong> organ <strong>of</strong>Corti.The cochlear duct communicates with <strong>the</strong> vestibularendolymph-containing sacs through two fine canalsso that <strong>the</strong> endolymphatic system <strong>of</strong> <strong>the</strong> cochlea<strong>and</strong> vestibule is continuous, like <strong>the</strong> perilymphaticone. Gravitational acceleration <strong>of</strong> <strong>the</strong> head is detectedin a sensory organ arranged within endolymph-containingsacs in <strong>the</strong> vestibule (<strong>the</strong> utricle <strong>and</strong> saccule),<strong>and</strong> angular acceleration is detected within tubes emanatingin three dimensions from <strong>the</strong> utricle (lateral,posterior <strong>and</strong> superior semicircular canals). The sensorycells are located as a thickened portion <strong>of</strong> epi<strong>the</strong>lium,<strong>the</strong> macula, in <strong>the</strong> saccule <strong>and</strong> utricle <strong>and</strong> araised prominence <strong>of</strong> epi<strong>the</strong>lium, <strong>the</strong> crista, in expansions<strong>of</strong> each semicircular canal, <strong>the</strong> ampullae. Thevestibular aqueduct contains <strong>the</strong> endolymphatic duct<strong>and</strong> sac, which constitute a blind <strong>of</strong>fshoot <strong>of</strong> <strong>the</strong> endolymphaticsystem, probably functioning to absorbendolymph. The cochlear aqueduct is a communicationbetween <strong>the</strong> cerebrospinal fluid in <strong>the</strong> subarachnoidspace to <strong>the</strong> perilymph <strong>of</strong> <strong>the</strong> scala tympani near<strong>the</strong> round window. Cochlea, vestibule <strong>and</strong> semicircularcanals are surrounded by very dense bone, <strong>the</strong> oticcapsule.The cochlear <strong>and</strong> vestibular sensory structuresare supplied by a double nerve, <strong>the</strong> audiovestibularnerve or eighth cranial nerve, which enters <strong>the</strong> temporalbone through <strong>the</strong> internal auditory meatus. Thefacial nerve or seventh cranial nerve, enters <strong>the</strong> temporalbone through <strong>the</strong> same canal, <strong>and</strong> after a rightangledbend in <strong>the</strong> genu, where <strong>the</strong> geniculate ganglionis located, reaches <strong>the</strong> posterior wall <strong>of</strong> <strong>the</strong> middleear, from which it passes down through <strong>the</strong> mastoid toemerge in <strong>the</strong> region <strong>of</strong> <strong>the</strong> parotid salivary gl<strong>and</strong>, afterwhich it provides motor nerve supply for <strong>the</strong> muscles<strong>of</strong> <strong>the</strong> face.8.1.3 HistologyThe ear <strong>and</strong> temporal bone are composed <strong>of</strong> many differenttissues, <strong>the</strong> normal histology <strong>of</strong> which is best consideredwhen describing <strong>the</strong> pathologic appearances <strong>of</strong><strong>the</strong>se parts.8.2 External Ear<strong>and</strong> Auditory Canal8.2.1 Inflammatory<strong>and</strong> Metabolic Lesions8.2.1.1 Diffuse External OtitisThis is a common condition, most frequently associatedwith Pseudomonas aeruginosa infection, but localtrauma is also an important causative factor. Histologicalfeatures include acute inflammation <strong>of</strong> <strong>the</strong> dermis,toge<strong>the</strong>r with acanthosis <strong>and</strong> hyperkeratosis <strong>of</strong> <strong>the</strong> epidermis.8.2.1.2 PerichondritisPerichondritis most commonly affects <strong>the</strong> pinna, whereit may follow surgical trauma. As in <strong>the</strong> diffuse acute inflammation<strong>of</strong> <strong>the</strong> ear canal, Pseudomonas aeruginosa is<strong>the</strong> most common infecting organism. Pus accumulatesbetween <strong>the</strong> perichondrium <strong>and</strong> cartilage <strong>of</strong> <strong>the</strong> pinna.This may interfere with <strong>the</strong> blood supply <strong>of</strong> <strong>the</strong> cartilage<strong>and</strong> so lead to its necrosis.8.2.1.3 Malignant Otitis ExternaMalignant otitis externa was first described as a severeinfection <strong>of</strong> <strong>the</strong> external auditory canal [16]. It usually(but not always [101]) affects elderly diabetics, resultingin unremitting pain, purulent discharge <strong>and</strong> invasion <strong>of</strong>cartilage, nerve, bone <strong>and</strong> adjacent s<strong>of</strong>t tissue. The causativeagent is usually Pseudomonas aeruginosa, but o<strong>the</strong>rorganisms including fungi have been incriminated. Thecondition frequently goes on to ninth, tenth, eleventh<strong>and</strong> twelfth cranial nerve palsies <strong>and</strong> meningitis <strong>and</strong>death may result.Histopathological changes in <strong>the</strong> temporal bones <strong>of</strong>two patients who had been diagnosed clinically as havingmalignant otitis externa <strong>and</strong> were thought to havedied <strong>of</strong> this condition [123] were those <strong>of</strong> severe otitismedia <strong>and</strong> osteomyelitis <strong>of</strong> <strong>the</strong> jugular foramen secondaryto it. It seems likely that <strong>the</strong> manifestations <strong>of</strong> malignantotitis media are due to <strong>the</strong> spread <strong>of</strong> inflammationfrom <strong>the</strong> tympanic cavity <strong>and</strong> mastoid air spaces to<strong>the</strong> petrous apex through bone marrow spaces by a process<strong>of</strong> osteomyelitis [85]. In recent years several patientswith AIDS have been reported to have malignant otitisexterna, <strong>and</strong> in one <strong>of</strong> <strong>the</strong>m <strong>the</strong> presence <strong>of</strong> acute osteomyelitis<strong>of</strong> <strong>the</strong> skull base in addition supported <strong>the</strong> concept<strong>of</strong> osteomyelitis as <strong>the</strong> pathologic basis for malignantotitis externa put forward above [121].


238 L. Michaels8.2.1.4 Relapsing Polychondritis8Relapsing polychondritis is a rare disease <strong>of</strong> unknownaetiology characterised by recurring bouts <strong>of</strong> inflammationaffecting cartilaginous structures <strong>and</strong> <strong>the</strong> eye.Although <strong>the</strong> cartilage <strong>of</strong> <strong>the</strong> external ear is most frequentlyinvolved <strong>and</strong> that <strong>of</strong> <strong>the</strong> nose next in frequency,it is <strong>the</strong> inflammation with destruction <strong>of</strong> <strong>the</strong> cartilages<strong>of</strong> <strong>the</strong> respiratory tract, particularly those <strong>of</strong> <strong>the</strong> larynx,which threatens life, <strong>and</strong> in most cases where death hasresulted from <strong>the</strong> condition it is from respiratory obstructiondue to such cartilage damage.The cartilage <strong>of</strong> <strong>the</strong> pinna, is recurrently acutely inflamed<strong>and</strong> this leads eventually to a cobblestone appearanceresembling boxer’s ear. Inflammation <strong>of</strong> <strong>the</strong>nasal septum leads to a sinking <strong>of</strong> this structure, producinga “saddle nose” appearance. Involvement <strong>of</strong> laryngeal<strong>and</strong> tracheal cartilages may lead to respiratorydifficulty. Rib cartilages may be swollen <strong>and</strong> tender.Episcleritis or scleritis, iritis, conjunctivitis or keratitismay also be found in relapsing polychondritis. Heart lesionsare characteristically aortic, showing signs <strong>of</strong> regurgitation.The histology <strong>of</strong> this affliction is dealt within Chap. 7.8.2.1.5 GoutGout is manifested both as an acute arthritis that is relatedto deposits <strong>of</strong> urates in <strong>the</strong> joint capsule, most frequentlyin <strong>the</strong> big toe joint, <strong>and</strong> as tophi in non-articulartissues. The external ear is one <strong>of</strong> <strong>the</strong> most frequentplaces for <strong>the</strong> latter <strong>and</strong> deposits may occur in <strong>the</strong> helix<strong>and</strong> antihelix. They may ulcerate, discharging a creamywhite material within which needle-like crystals <strong>of</strong> sodiumurate may be detected on microscopy. For histology,see Chap. 7.8.2.1.6 OchronosisOchronosis (alkaptonuria) is an inherited disease <strong>of</strong> <strong>the</strong>metabolism in which a step in tyrosine metabolism isdisturbed, resulting in accumulation <strong>of</strong> homogentisicacid in a variety <strong>of</strong> places, but especially in <strong>the</strong> cartilages.The substance is colourless in <strong>the</strong> urine whenfirst passed, but darkens to a black or brown polymeron st<strong>and</strong>ing. The disease is inherited as an autosomalrecessive.In <strong>the</strong> external ear <strong>the</strong>re may be one or both <strong>of</strong> twomanifestations: (a) dark colour <strong>of</strong> <strong>the</strong> wax; when seenin a child this may be <strong>the</strong> first manifestation <strong>of</strong> ochronosis;(b) dark colour <strong>of</strong> <strong>the</strong> aural cartilage due to <strong>the</strong>binding <strong>of</strong> <strong>the</strong> homogentisic acid to <strong>the</strong> cartilage groundsubstance.Fig. 8.2. Idiopathic pseudocystic chondromalacia <strong>of</strong> <strong>the</strong> cartilage<strong>of</strong> <strong>the</strong> auricle. Note <strong>the</strong> cystic cavity in <strong>the</strong> cartilage lined with degeneratedcartilage that has lost its elastic tissue. Normal elastictissue is present fur<strong>the</strong>r away from <strong>the</strong> pseudocyst at <strong>the</strong> left h<strong>and</strong>corner. Reproduced from Michaels <strong>and</strong> Hellquist [68]8.2.2 Pseudocystic<strong>and</strong> Cystic Lesions8.2.2.1 Idiopathic PseudocysticChondromalaciaIdiopathic pseudocystic chondromalacia occurs mainlyin young <strong>and</strong> middle-aged adults. The gross appearanceis one <strong>of</strong> a localised swelling <strong>of</strong> <strong>the</strong> auricular cartilage. Acut surface shows a well-defined cavity in <strong>the</strong> cartilage,which is distended with yellowish watery fluid [42]. Microscopically,<strong>the</strong> cavity shows a lining <strong>of</strong> degeneratedcartilage on one surface (Fig. 8.2); on <strong>the</strong> o<strong>the</strong>r surface<strong>the</strong> cartilage is normal. It seems possible that <strong>the</strong> fluid isan exudate from undamaged perichondrial vessels thatcannot be absorbed by <strong>the</strong> damaged perichondrial vessels.The association <strong>of</strong> this lesion with severe atopic eczemain four children [27] suggested that minor traumafrom repeated rubbing <strong>of</strong> <strong>the</strong> auricle may play a part.Small pseudocysts <strong>of</strong> <strong>the</strong> elastic cartilage <strong>of</strong> <strong>the</strong> pinnamay also be seen in <strong>the</strong> vicinity <strong>of</strong> inflammatory or neoplasticlesions <strong>of</strong> that region.8.2.2.2 First Branchial Cleft CystA developmental origin from <strong>the</strong> first branchial cleft hasbeen assumed in some fistulas, sinuses, <strong>and</strong> cysts arisingin <strong>the</strong> periauricular <strong>and</strong> <strong>the</strong> parotid region <strong>and</strong> also insome fistulas arising in <strong>the</strong> neck that are located abovea horizontal plane passing through <strong>the</strong> hyoid bone. Thecommonest <strong>of</strong> <strong>the</strong>se lesions is <strong>the</strong> preauricular sinus.This usually shows a stratified squamous epi<strong>the</strong>lial lining,but occasionally it may be <strong>of</strong> respiratory epi<strong>the</strong>lium,


Ear <strong>and</strong> Temporal Bone Chapter 8 239deep to which <strong>the</strong> connective tissue is chronically inflamed.There is <strong>of</strong>ten elastic cartilage in <strong>the</strong> deep wall<strong>of</strong> <strong>the</strong> sinus. Branchial cleft cysts are more extensivelydiscussed in Chap. 9.8.2.3 Tumour-Like Lesions8.2.3.1 ChondrodermatitisNodularis HelicisIn chondrodermatitis nodularis chronica helicis, sometimesknown as Winkler’s disease, a small nodule formson <strong>the</strong> auricle, usually in <strong>the</strong> superior portion <strong>of</strong> <strong>the</strong> helix.Pain is always a prominent feature.Histological examination <strong>of</strong> biopsies <strong>of</strong> such lesionsin which <strong>the</strong> elastic cartilage underlying <strong>the</strong> skin <strong>of</strong> <strong>the</strong>auricle is particularly well-represented, shows ulceration<strong>of</strong> <strong>the</strong> skin <strong>of</strong> <strong>the</strong> auricle <strong>and</strong> complete necrosis <strong>of</strong> <strong>the</strong> tip<strong>of</strong> <strong>the</strong> underlying elastic cartilage <strong>of</strong> <strong>the</strong> auricle. In somecases a piece <strong>of</strong> extruded necrotic cartilage may be seenon <strong>the</strong> floor <strong>of</strong> <strong>the</strong> ulcer. The perichondrium <strong>of</strong> <strong>the</strong> elasticcartilage in this region shows obstructive thickening<strong>of</strong> small arteries.The helix is one <strong>of</strong> <strong>the</strong> fur<strong>the</strong>st points from <strong>the</strong> source<strong>of</strong> <strong>the</strong> arterial blood supply <strong>of</strong> <strong>the</strong> pinna. It seems likelythat obstruction <strong>of</strong> small arteries <strong>of</strong> <strong>the</strong> perichondriumis <strong>the</strong> primary lesion leading to cartilage necrosis,<strong>and</strong> that <strong>the</strong> acute inflammation <strong>and</strong> epidermal ulcerationare secondary to <strong>the</strong> nearby cartilage necrosis. Anassociation between chondrodermatitis nodularis helicis<strong>and</strong> systemic sclerosis has been described [12]. In thiscondition obstructive changes are frequently found insmall arteries.8.2.3.2 Keratosis Obturans<strong>and</strong> Cholesteatoma<strong>of</strong> External CanalIn keratosis obturans <strong>the</strong> keratin produced by exfoliationfrom <strong>the</strong> skin <strong>of</strong> <strong>the</strong> tympanic membrane <strong>and</strong> externalcanal is retained on <strong>the</strong> epi<strong>the</strong>lial surface <strong>and</strong> forms asolid plug. This enlarges <strong>and</strong> may cause circumferentialerosion <strong>of</strong> <strong>the</strong> bony canal. Keratosis obturans is probably<strong>the</strong> result <strong>of</strong> a defect <strong>of</strong> <strong>the</strong> normal migratory properties<strong>of</strong> <strong>the</strong> squamous epi<strong>the</strong>lium <strong>of</strong> tympanic membrane<strong>and</strong> adjacent ear canal that causes <strong>the</strong> accumulation <strong>of</strong>keratinous debris [20]. A minor degree <strong>of</strong> this process –keratosis <strong>of</strong> <strong>the</strong> tympanic membrane – in which deposits<strong>of</strong> keratin grow on <strong>the</strong> eardrum <strong>and</strong> cause tinnitus hasalso been found to be associated with absent or defectiveauditory epi<strong>the</strong>lial migration [103].A condition that has been distinguished from keratosisobturans is cholesteatoma <strong>of</strong> <strong>the</strong> external canal. Inthis lesion epidermoid tissue appears to penetrate into<strong>the</strong> wall <strong>of</strong> <strong>the</strong> deep external canal causing localised osteonecrosis<strong>and</strong> bone erosion [87].8.2.3.3 Keratin GranulomaA granulomatous process may result in <strong>the</strong> external earcanal when keratin squames become implanted into <strong>the</strong>deeper tissues following traumatic laceration [39]. Thegranuloma contains foreign body-type giant cells, histiocytes,lymphocytes, plasma cells <strong>and</strong> flakes <strong>of</strong> keratin.The latter are strongly eosinophilic <strong>and</strong> birefringentin polarised light. Aural polyps frequently contain suchgranulomas, but <strong>the</strong> keratin is <strong>the</strong>n more likely to be derivedfrom a middle ear cholesteatoma (see below).8.2.3.4 Angiolymphoid Hyperplasiawith Eosinophilia<strong>and</strong> Kimura’s DiseaseSynonyms for angiolymphoid hyperplasia with eosinophiliaare epi<strong>the</strong>lioid haemangioma, benign angiomatousnodules <strong>of</strong> face <strong>and</strong> scalp, atypical pyogenic granuloma,<strong>and</strong> several o<strong>the</strong>r terms. Although this entity wasfirst described by Kimura, “Kimura’s disease” is nowbelieved to be a different condition (see below).Angiolymphoid hyperplasia with eosinophilia mayoccur anywhere in <strong>the</strong> skin, especially on <strong>the</strong> scalp <strong>and</strong>face, but <strong>the</strong>re is a particular predilection for <strong>the</strong> externalauricle <strong>and</strong> external auditory meatus. It is a lesion <strong>of</strong>young <strong>and</strong> middle-aged <strong>of</strong> both sexes <strong>and</strong> all races.Grossly, <strong>the</strong>re are sessile or plaque-like red or reddish-bluelesions from 2 to 10 mm in diameter, whichmay coalesce to form large plaques that obstruct <strong>the</strong> earcanal. On transection <strong>the</strong> lesion is seen to be presentin <strong>the</strong> dermis <strong>and</strong> subcutaneous tissue. Microscopically,<strong>the</strong>re is a mixture <strong>of</strong> two proliferated elements in <strong>the</strong>dermis: blood vessels <strong>and</strong> lymphoid tissue. The bloodvessels are mainly capillaries lined by plump, <strong>of</strong>ten protruding(hobnailed) sometimes multilayered, endo<strong>the</strong>lialcells (Fig. 8.3). Occasionally an artery or vein showingintimal fibrous thickening is part <strong>of</strong> <strong>the</strong> vascular component.Solid clusters <strong>of</strong> cells, which are <strong>of</strong>ten vacuolated,show features intermediate between endo<strong>the</strong>lial cells<strong>and</strong> histiocytes are also observed [6]. The lymphoid tissuemay possess germinal centres. Eosinophils (<strong>of</strong>tenextremely numerous), mast cells <strong>and</strong> macrophages mayalso be prominent.Kimura’s disease is commoner in orientals, mainlyaffecting young males. It is a chronic inflammatory condition<strong>of</strong> unknown aetiology. It presents as large, deep<strong>and</strong> <strong>of</strong>ten disfiguring, subcutaneous masses in <strong>the</strong> preauricular,parotid <strong>and</strong> subm<strong>and</strong>ibular regions. Often,<strong>the</strong>re is enlargement <strong>of</strong> regional lymph nodes. Occasion-


240 L. Michaels<strong>of</strong> <strong>the</strong> oculoauriculovertebral syndrome (Goldenhar’ssyndrome) [49]. The lesion, which may be bilateral, isusually situated anterior to <strong>the</strong> normal tragus. Like <strong>the</strong>latter, <strong>the</strong> accessory tragus is composed <strong>of</strong> elastic cartilagewith a covering <strong>of</strong> skin. In rare cases a similar structurehas been found in <strong>the</strong> neck [8] or in <strong>the</strong> suprasternalregion [54].8Fig. 8.3. Epi<strong>the</strong>lioid haemangioma showing “hobnailed” endo<strong>the</strong>lium<strong>of</strong> <strong>the</strong> capillary on <strong>the</strong> left <strong>and</strong> lymphocytic infiltrationally, only lymph nodes are involved. There is a peripheralblood eosinophilia <strong>and</strong> raised levels <strong>of</strong> IgE.Microscopically, <strong>the</strong> subcutaneous masses are foundto be composed <strong>of</strong> lymphoid follicles surrounded by oedematousconnective tissue rich in eosinophils <strong>and</strong> containingnumerous thin-walled blood vessels resemblinghigh endo<strong>the</strong>lial venules [6]. Infiltration <strong>of</strong> <strong>the</strong> germinalcentres with eosinophils <strong>and</strong> follicle lysis is a frequentfinding, as is <strong>the</strong> presence <strong>of</strong> multinucleated cells. Thereis deposition <strong>of</strong> IgE on <strong>the</strong> processes <strong>of</strong> <strong>the</strong> folliculardendritic cells <strong>and</strong> <strong>the</strong>re are also numerous mast cells,<strong>the</strong> latter well shown by immunostaining with IgE mAb.Plasma cells may also be prominent.Diagnostic confusion between angiolymphoid hyperplasiawith eosinophilia <strong>and</strong> Kimura’s disease is likelyto occur if <strong>the</strong> characteristic vascular appearances inangiolymphoid hyperplasia with eosinophilia have notbeen recognised <strong>and</strong> if <strong>the</strong>re is very prominent lymphoidhyperplasia with follicle formation <strong>and</strong> markedtissue eosinophilia in <strong>the</strong> latter. In Kimura’s disease <strong>the</strong>endo<strong>the</strong>lial cells in <strong>the</strong> proliferating vessels never showan epi<strong>the</strong>lioid/histiocytoid appearance, nor is <strong>the</strong>re anassociated large vessel with intimal thickening. In angiolymphoidhyperplasia with eosinophilia <strong>the</strong>re is neverlymph node enlargement or tissue deposition <strong>of</strong> IgE.Both epi<strong>the</strong>lioid haemangioma <strong>and</strong> Kimura’s diseaseare benign entities. Recurrence is rare in <strong>the</strong> former ifit is completely excised. It is more frequent in <strong>the</strong> latter,but eventually it becomes stationary. A nephrotic syndromemay rarely occur in Kimura’s disease.8.2.3.5 Accessory TragusAccessory tragus is a fairly common congenital malformation<strong>of</strong> <strong>the</strong> external ear. In <strong>the</strong> majority <strong>of</strong> cases itis an isolated developmental defect not associated witho<strong>the</strong>r abnormalities. It is, however, a consistent feature8.2.3.6 KeloidKeloid, a common benign skin lesion, follows injury to<strong>the</strong> skin <strong>of</strong> <strong>the</strong> ear, <strong>of</strong>ten after piercing <strong>the</strong> earlobes forwearing an ear-ring. It is particularly frequent in blackpeople. Grossly, <strong>the</strong>re is a lobulated swelling covered innormal skin. Microscopically, <strong>the</strong> dermis is enlarged bydeposits <strong>of</strong> eosinophilic, poorly cellular collagen.8.2.4 Benign NeoplasmsExternal ear neoplasms derived from ceruminal gl<strong>and</strong>sare very uncommon. Only <strong>the</strong> adenoma can usually becategorised with certainty as being derived specificallyfrom ceruminous gl<strong>and</strong>s, since its component acini displayan apocrine secretory structure. Syringocystadenomapapilliferum <strong>and</strong> adenoid cystic carcinoma arisingin this region can sometimes appear to be developingfrom ceruminous gl<strong>and</strong>s.8.2.4.1 Adenoma <strong>of</strong> Ceruminal Gl<strong>and</strong>sICD-O:8420/0Adenomas are unusual neoplasms that present with ablockage <strong>of</strong> <strong>the</strong> lateral part <strong>of</strong> <strong>the</strong> external auditory meatus,<strong>of</strong>ten associated with deafness <strong>and</strong> discharge. Animportant part <strong>of</strong> <strong>the</strong> clinical investigation <strong>of</strong> all gl<strong>and</strong>ularneoplasms <strong>of</strong> <strong>the</strong> ear canal is exclusion <strong>of</strong> an originin <strong>the</strong> parotid gl<strong>and</strong>.Gross appearances are those <strong>of</strong> a superficial greymass up to 4 cm in diameter, which is covered in skin.Microscopically this neoplasm lacks a definite capsule.It is composed <strong>of</strong> regular gl<strong>and</strong>s <strong>of</strong>ten with intraluminalprojections (Fig. 8.4). The gl<strong>and</strong>ular epi<strong>the</strong>lium isbilayered, <strong>the</strong> outer layer being myoepi<strong>the</strong>lial, but thismay not be obvious in all parts <strong>of</strong> <strong>the</strong> neoplasm. Thegl<strong>and</strong>s are <strong>of</strong>ten arranged in groups surrounded by fibroustissue. In some ceruminomas, acid-fast fluorescentpigment may be found in <strong>the</strong> tumour cells that issimilar to that found in normal ceruminal gl<strong>and</strong>s [15,125].Adenoma <strong>of</strong> ceruminal gl<strong>and</strong>s is a benign neoplasm.Recurrence should not be expected if it is carefully excised.


Ear <strong>and</strong> Temporal Bone Chapter 8 241Fig. 8.4. Ceruminoma <strong>of</strong> <strong>the</strong> external canal. Note peg-like protrusionsinto <strong>the</strong> lumina, indicating apocrine secretion. Reproducedfrom Michaels <strong>and</strong> Hellquist [68]Fig. 8.5. Osteoma <strong>of</strong> <strong>the</strong> deep external canal. Reproduced fromMichaels <strong>and</strong> Hellquist [68]8.2.4.2 Pleomorphic Adenoma<strong>of</strong> Ceruminal Gl<strong>and</strong>sICD-O:8940/0A benign neoplasm <strong>of</strong> <strong>the</strong> skin with a structure similarto that <strong>of</strong> pleomorphic adenoma <strong>of</strong> salivary gl<strong>and</strong>s isalso occasionally seen in <strong>the</strong> external auditory meatus.Cartilage, myoepi<strong>the</strong>lial <strong>and</strong> adenomatous structuresare features <strong>of</strong> this neoplasm.8.2.4.3 SyringocystadenomaPapilliferum<strong>of</strong> Ceruminal Gl<strong>and</strong>sICD-O:8406/0Syringocystadenoma papilliferum is seen in children oryoung adults usually on <strong>the</strong> scalp or face. Occasionallyit occurs in <strong>the</strong> ear canal. The histological appearance<strong>of</strong> <strong>the</strong> neoplasm is that <strong>of</strong> an invagination from <strong>the</strong> surfaceepi<strong>the</strong>lium forming a cyst-like structure. Projectinginto <strong>the</strong> lumen are papillae lined with bilayered gl<strong>and</strong>ularepi<strong>the</strong>lium, which may show decapitation secretiontypical <strong>of</strong> apocrine (ceruminal) gl<strong>and</strong>s.8.2.4.4 Bony LesionsBecause <strong>of</strong> <strong>the</strong> difficulty <strong>of</strong> classifying a solitary benignneoplasm composed <strong>of</strong> woven bone <strong>and</strong> fibrous tissueinto one or o<strong>the</strong>r <strong>of</strong> <strong>the</strong> classical groups – monostoticfibrous dysplasia or ossifying fibroma – <strong>the</strong> designationbenign fibro-osseous lesion (fibrous dysplasia) maybe used in most circumstances without loss <strong>of</strong> accuracy.Lesions <strong>of</strong> this type are found in <strong>the</strong> temporal bone <strong>of</strong>tenpresenting deep to <strong>the</strong> external ear [65, 80]. On o<strong>the</strong>rsides, this lumping is less appropriate, as discussed inChap. 4.The main clinical features are progressive loss <strong>of</strong>hearing, conductive in most, sensorineural, which canbe pr<strong>of</strong>ound, in some <strong>and</strong> enlargement <strong>of</strong> <strong>the</strong> temporalbone with progressive bony occlusion <strong>of</strong> <strong>the</strong> external auditorymeatus. Facial nerve palsy is present in some patientsdue to involvement <strong>of</strong> <strong>the</strong> facial nerve by <strong>the</strong> pathologicalprocess.The gross appearance <strong>of</strong> benign fibro-osseous lesionsis one <strong>of</strong> yellowish-white resilient tissue, whichoccasionally includes small cysts filled with an ambercolouredfluid. The transition to normal bone is sharp.Microscopically, irregular trabeculae <strong>of</strong> woven bone areembedded in a connective tissue stroma. The constriction<strong>of</strong> <strong>the</strong> ear canal may cause an epidermoid cyst lateralto <strong>the</strong> tympanic membrane, referred to in some publicationsas “cholesteatoma” [65].Although fibrous dysplasia has been on rare occasionsassociated with malignant disease such as osteogenicsarcoma, fibrosarcoma, chondrosarcoma <strong>and</strong> giantcell tumour, <strong>the</strong> temporal bone is not one <strong>of</strong> <strong>the</strong> siteswhere this change has been described.Osteoma <strong>and</strong> exostosis are two types <strong>of</strong> benign bonyenlargement <strong>of</strong> <strong>the</strong> deeper bony portion <strong>of</strong> <strong>the</strong> externalauditory canal. Osteoma is a spherical mass arising from<strong>the</strong> region <strong>of</strong> <strong>the</strong> tympanosquamous or tympanomastoidsuture line by a distinct bony pedicle. Symptoms areusually those <strong>of</strong> ear canal obstruction. Microscopically,<strong>the</strong> osteoma is composed <strong>of</strong> lamellar bone <strong>and</strong> may showouter cortical <strong>and</strong> inner cancellous trabeculated areas,<strong>the</strong> latter with marrow spaces. There may be a thin layer<strong>of</strong> woven bone on <strong>the</strong> surfaces <strong>of</strong> <strong>the</strong> lamellar bone. Theosteoma is covered in <strong>the</strong> normal squamous epi<strong>the</strong>lium<strong>of</strong> <strong>the</strong> ear canal (Fig. 8.5).Exostosis is a broad-based lesion, which is <strong>of</strong>ten bilateral<strong>and</strong> symmetrical. It is usually situated deeperin <strong>the</strong> ear canal than osteomas. In <strong>the</strong> bony portion<strong>of</strong> <strong>the</strong> normal external auditory meatus <strong>the</strong>re are noadnexal structures, <strong>and</strong> <strong>the</strong> subcutaneous tissue <strong>and</strong>


242 L. Michaels8.2.5 Malignant Neoplasms8Fig. 8.6. Exostosis <strong>of</strong> <strong>the</strong> deep external canal. Note thin epidermallayer on <strong>the</strong> exostosis <strong>and</strong> canal skin <strong>and</strong> <strong>the</strong>ir proximity to<strong>the</strong> bone. Reproduced from Michaels <strong>and</strong> Hellquist [68]periosteum merge to form a thin layer. The distancebetween <strong>the</strong> epidermal surface <strong>and</strong> underlying bone isconsequently small. This explains <strong>the</strong> propensity towardsexostoses <strong>of</strong> <strong>the</strong> tympanic bone to develop inthis region in those who swim frequently in cold water.It seems likely that <strong>the</strong> water, after dribbling into<strong>the</strong> deep external auditory canal, exerts a cooling effecton <strong>the</strong> bone surface <strong>and</strong> stimulates it to producenew bone. Unlike osteoma (Fig. 8.5) <strong>the</strong> bone formations<strong>of</strong> exostosis are said not to possess any marrowspaces (Fig. 8.6).Osteoma <strong>and</strong> exostosis are <strong>of</strong>ten associated with infection<strong>of</strong> <strong>the</strong> external canal on <strong>the</strong>ir tympanic membraneside <strong>and</strong> surgical removal may be required to enhancedrainage as well as to relieve <strong>the</strong> conductive hearingloss.Five cases <strong>of</strong> a benign circumscribed bony lesion <strong>of</strong><strong>the</strong> external auditory canal distinct from exostosis <strong>and</strong>osteoma have recently been described [91]. They allshowed a hard, round, unilateral, skin-covered mass occluding<strong>the</strong> superficial external auditory canal with norelationship to <strong>the</strong> cartilaginous tissue or to <strong>the</strong> bonystructure surrounding that canal. Histologically, <strong>the</strong>lesion displayed an osteoma-like bone formation withsparse osteoblastic areas; mature lamellar bone was observedsome cases, <strong>and</strong> also bone marrow containingadipose tissue <strong>and</strong> hematopoietic remnants. The boneshowed irregular trabeculae, bordered by osteoid osteoblasts.8.2.5.1 Adenocarcinoma<strong>of</strong> Ceruminal Gl<strong>and</strong>sICD-O:8420/3Adenocarcinoma <strong>of</strong> ceruminal gl<strong>and</strong>s is a rare neoplasmpresenting in <strong>the</strong> superficial part <strong>of</strong> <strong>the</strong> external ear canal.There is always local infiltration. The neoplasmpossesses a gl<strong>and</strong>ular structure with evidence <strong>of</strong> apocrinedifferentiation, but <strong>the</strong> gl<strong>and</strong>s show loss <strong>of</strong> a myoepi<strong>the</strong>liallayer <strong>and</strong> <strong>the</strong> cells are markedly atypical withincreased mitotic activity. Recurrence is to be expectedfollowing surgical removal. Death due to involvement <strong>of</strong>local vital structures has been reported. Rare examples<strong>of</strong> low-grade adenocarcinoma <strong>of</strong> ceruminal gl<strong>and</strong>s havebeen documented.8.2.5.2 Adenoid Cystic Carcinoma<strong>of</strong> Ceruminal Gl<strong>and</strong>sICD-O:8200/3This malignant neoplasm has <strong>the</strong> gross <strong>and</strong> microscopicfeatures <strong>of</strong> <strong>the</strong> corresponding major or minor salivarygl<strong>and</strong> neoplasm, including its tendency to invade alongnerve sheaths. Relentless though <strong>of</strong>ten delayed recurrence<strong>and</strong> eventual bloodstream metastasis, particularlyto <strong>the</strong> lungs, are likewise features <strong>of</strong> this cancer.8.2.5.3 Basal Cell CarcinomaICD-O:8090/3The great majority <strong>of</strong> malignant epi<strong>the</strong>lial neoplasms<strong>of</strong> <strong>the</strong> pinna are basal cell carcinomas, a small numberonly being squamous cell carcinomas. The few basal cellcarcinomas that occur in <strong>the</strong> ear canal arise near <strong>the</strong> externalopening. Their preference for <strong>the</strong> exposed part <strong>of</strong><strong>the</strong> external ear is in keeping with <strong>the</strong> accepted view thatsunlight is in most cases <strong>the</strong> causal factor in skin insufficientlyprotected by melanin pigment.The gross appearance <strong>of</strong> basal cell carcinoma is usuallyone <strong>of</strong> a pearly wax-like nodule that eventually ulcerates.Twenty-five per cent <strong>of</strong> basal cell carcinomas <strong>of</strong><strong>the</strong> pinna are <strong>of</strong> <strong>the</strong> morphea type (see below). The importance<strong>of</strong> this variety is that although <strong>the</strong> edge <strong>of</strong> <strong>the</strong>tumour tends to infiltrate subcutaneously, this cannotbe recognised clinically or on gross pathological examination.The classical <strong>and</strong> most frequent form <strong>of</strong> basalcell carcinoma is composed <strong>of</strong> solid masses <strong>of</strong> cells,which are seen to be arising from <strong>the</strong> basal layers <strong>of</strong> <strong>the</strong>epidermis or <strong>the</strong> outer layers <strong>of</strong> <strong>the</strong> hair follicles. Thecells are uniform with basophilic nuclei <strong>and</strong> little cy-


Ear <strong>and</strong> Temporal Bone Chapter 8 243Fig. 8.7. Morphea type <strong>of</strong> basal cell carcinoma showing thindowngrowths with stroma <strong>of</strong> inflammatory connective tissue. Reproducedfrom Michaels <strong>and</strong> Hellquist [68]toplasm. At <strong>the</strong> periphery <strong>of</strong> <strong>the</strong> neoplastic lobules <strong>the</strong>cells tend to be palisaded. Mitoses are frequent as are alveolaror cystic spaces. Squamous cell differentiation isalso common.The splitting up <strong>of</strong> cell groups by much hyaline fibroustissue, so that <strong>the</strong> carcinoma appears compressedinto thin str<strong>and</strong>s, is referred to as <strong>the</strong> morphea type <strong>of</strong>basal cell carcinoma (Fig. 8.7). The suggestion that tumourswith this histology have a worse outlook is probablyrelated to <strong>the</strong>ir tendency towards insidious infiltration(see above). There is o<strong>the</strong>rwise no convincingevidence <strong>of</strong> <strong>the</strong> relationship <strong>of</strong> a particular histologicalappearance to prognosis in basal cell carcinoma.However, when immunohistochemical assessment forKi-67 antigen (MIB1 in paraffin sections), a proliferation-associatedantigen, is performed on basal cell carcinomas,those tumours that recur have been shown topossess a higher proportion <strong>of</strong> cells positive for that antigenthan those that do not [40]. The degree <strong>of</strong> tumourangiogenesis is ano<strong>the</strong>r histologic factor that showspromise in judging <strong>the</strong> prognosis <strong>of</strong> basal cell carcinoma[107].This is not an aggressive neoplasm <strong>and</strong> in at least 90%<strong>of</strong> cases a 3-year cure can be easily achieved by surgicalexcision. In a few cases repeated recurrences with deepextension to <strong>the</strong> middle ear, mastoid <strong>and</strong> even cranialcavity may, however, take place. Metastasis is rare.Pinna lesions in a prominent position are identifiedearly. A serious problem with <strong>the</strong> canal lesions is <strong>the</strong> delayin diagnosis because <strong>of</strong> <strong>the</strong> minimal symptoms thatmay be present. Pain, hearing loss <strong>and</strong> drainage <strong>of</strong> bloodor pus are <strong>the</strong> main features in that group. A plaque-likeor even polypoid mass may be felt or even seen.Squamous carcinomas arising on <strong>the</strong> pinna grosslyresemble those seen elsewhere on <strong>the</strong> skin. The appearances<strong>of</strong> <strong>the</strong> canal lesions are those <strong>of</strong> a mass, sometimeswarty, occluding <strong>the</strong> lumen <strong>and</strong> invading deeply into<strong>the</strong> surrounding tissues. There may be dissolution <strong>of</strong> <strong>the</strong>tympanic membrane with invasion <strong>of</strong> <strong>the</strong> middle ear.Squamous cell carcinoma <strong>of</strong> <strong>the</strong> external ear usuallyshows significant degrees <strong>of</strong> keratinisation. In <strong>the</strong>cases with a canal origin evidence <strong>of</strong> origin from canalepidermis is usually present. In cases arising deepwithin <strong>the</strong> ear canal <strong>the</strong>re is usually a concomitant originfrom middle ear epi<strong>the</strong>lium <strong>and</strong> dissolution <strong>of</strong> <strong>the</strong>tympanic membrane (see below). The neoplasm may beso well differentiated that it can be confused with benignpapilloma. The association <strong>of</strong> a well-differentiatedsquamous carcinoma with marked desmoplasia mayalso delay <strong>the</strong> correct diagnosis. The verrucous form <strong>of</strong>squamous cell carcinoma has been seen in <strong>the</strong> externalear [105]. Metastatic spread <strong>of</strong> squamous carcinoma<strong>of</strong> <strong>the</strong> pinna <strong>and</strong> external auditory meatus to lymphnodes is unusual. Squamous carcinoma <strong>of</strong> <strong>the</strong> externalcanal is an aggressive disease with a high propensitytowards local recurrence. The outcome <strong>of</strong> <strong>the</strong> diseasefollowing surgical excision is related to <strong>the</strong> clinicalstage at presentation; <strong>the</strong> higher <strong>the</strong> stage <strong>the</strong> worse<strong>the</strong> outcome [83].8.2.5.5 Melanotic NeoplasmsMelanotic neoplasms are unusual in <strong>the</strong> external ear.Nevi arise mainly in <strong>the</strong> ear canal, but are rare on <strong>the</strong>auricle. Malignant melanomas, on <strong>the</strong> o<strong>the</strong>r h<strong>and</strong> usuallyarise on <strong>the</strong> auricle; origin in <strong>the</strong> external canal isextremely unusual [72]. Malignant melanoma <strong>of</strong> <strong>the</strong> externalear is a highly malignant disease. In a review <strong>of</strong> 16patients with this condition as many as 9 cases showedinvasion to Clark level IV or more [22]. It is likely thatcervical <strong>and</strong> parotid gl<strong>and</strong> lymph nodes will be involvedwhen malignant melanoma <strong>of</strong> <strong>the</strong> external ear is firstdiagnosed [99].8.2.5.4 Squamous Cell CarcinomaICD-O:8070/3The majority <strong>of</strong> squamous cell carcinomas <strong>of</strong> <strong>the</strong> externalear arise in <strong>the</strong> pinna; a lesser number arise in <strong>the</strong>external canal.


244 L. Michaels88.3 Middle Ear<strong>and</strong> Mastoid8.3.1 Inflammatory Lesions8.3.1.1 Acute<strong>and</strong> Chronic Otitis MediaOtitis media is one <strong>of</strong> <strong>the</strong> most common <strong>of</strong> all diseases,particularly in young children. The disease is usuallycaused by bacterial infection, Haemophilus influenzae<strong>and</strong> Gram-positive cocci usually being incriminated in<strong>the</strong> acute form <strong>and</strong> Gram-negative bacilli in <strong>the</strong> chronicform. The clinical forms <strong>of</strong> <strong>the</strong> acute <strong>and</strong> chronic conditionscorrespond to <strong>the</strong> pathological changes, but intermediateor mixed states are frequent. Perforation <strong>of</strong><strong>the</strong> tympanic membrane may occur at any phase <strong>of</strong> otitismedia, but an effusion, accompanied by all <strong>of</strong> <strong>the</strong> o<strong>the</strong>rmanifestations <strong>of</strong> chronic otitis media, is <strong>of</strong>ten presentbehind an intact tympanic membrane, a conditionknown as serous otitis media.The appearances <strong>of</strong> <strong>the</strong> middle ear mucosa in acute otitismedia may be seen in <strong>the</strong> bone chips removed at mastoidectomy.There is congestion <strong>and</strong> oedema <strong>of</strong> <strong>the</strong> mucosa<strong>of</strong> <strong>the</strong> mastoid air cells. Haemorrhage may be severe<strong>and</strong> <strong>the</strong> mucosa <strong>and</strong> air cells are filled with neutrophils.Pus destroys bone, <strong>the</strong> actual dissolution being carriedout by osteoclasts. At <strong>the</strong> same time new bone formationtakes place, commencing as osteoid, later becoming woven<strong>and</strong> finally lamellar. Fibrosis may also be active evenin <strong>the</strong> acute stage. Acute inflammatory changes are alsoprominent in o<strong>the</strong>r parts <strong>of</strong> <strong>the</strong> middle ear. The tympanicmembrane shows marked congestion, <strong>the</strong> dilated vesselsdistending <strong>the</strong> connective tissue layer. Pus cells fill <strong>the</strong>middle ear cavity. The acute inflammation may spreaddeep into <strong>the</strong> temporal bone as osteomyelitis.The chronic form <strong>of</strong> otitis media is associated withnecrosis, caused by <strong>the</strong> bacterial infection. There is, as in<strong>the</strong> acute form, marked congestion. The latter results inhaemorrhage in many cases. Because <strong>of</strong> <strong>the</strong> poor lymphdrainage in <strong>the</strong> middle ear old haematoma becomesconverted into cholesterol granuloma, with cholesterolclefts surrounded by foreign body-type giant cells, <strong>and</strong>haemosiderin.Associated with <strong>the</strong>se changes <strong>and</strong> representing animportant part <strong>of</strong> <strong>the</strong> pathological picture, is proliferativeactivity <strong>of</strong> middle ear tissue. The columnar epi<strong>the</strong>lium<strong>of</strong> <strong>the</strong> middle ear has, in <strong>the</strong> presence <strong>of</strong> inflammationor o<strong>the</strong>r pathological changes in <strong>the</strong> middle ear,<strong>the</strong> remarkable property <strong>of</strong> invaginating itself to producegl<strong>and</strong>s, which <strong>of</strong>ten develop luminal secretion.The gl<strong>and</strong>ular transformation <strong>of</strong> <strong>the</strong> middle ear mucosa,known as gl<strong>and</strong>ular metaplasia, may be seen in anypart <strong>of</strong> <strong>the</strong> cleft, including <strong>the</strong> mastoid ear cells. The secretion<strong>of</strong> <strong>the</strong> gl<strong>and</strong>s contributes to <strong>the</strong> exudate in otitismedia with effusion. Fibrous tissue proliferation mayalso occur in combination with gl<strong>and</strong>ular transformation– a process which, in <strong>the</strong> advanced state, has beencalled “fibrocystic sclerosis” [95].A specific form <strong>of</strong> reparative reaction following inflammationis <strong>the</strong> development <strong>of</strong> granulation tissue. Inthis process, <strong>the</strong> endo<strong>the</strong>lium <strong>of</strong> blood vessels <strong>and</strong> fibroblastsare <strong>the</strong> newly formed cells. Mononuclear inflammatorycells usually accompany <strong>the</strong> latter. The granulationtissue is usually particularly prominent in <strong>the</strong> middleear under <strong>the</strong> mucosa covering <strong>the</strong> promontory fromwhich it frequently protrudes into <strong>the</strong> external canalthrough a perforation <strong>of</strong> <strong>the</strong> tympanic membrane, formingan aural polyp that is covered in pseudostratified columnar,ciliated respiratory or stratified squamous epi<strong>the</strong>lium.Fibroblasts <strong>and</strong> collagen are abundant in <strong>the</strong>terminal phase <strong>of</strong> <strong>the</strong> reparative stage.A normal degree <strong>of</strong> fibroblast cellularity in <strong>the</strong> fibrousreaction is seen in adhesive otitis media. A peculiar form <strong>of</strong>scar tissue production occurs in <strong>the</strong> middle ear, in which<strong>the</strong> collagen is poorly cellular <strong>and</strong> hyalinised. This condition,known as tympanosclerosis, is also characterised bydeposition <strong>of</strong> calcium salts in <strong>the</strong> hyaline fibrous tissue.The bony walls <strong>of</strong> <strong>the</strong> middle ear also frequently react to<strong>the</strong> inflammatory process with a new formation <strong>of</strong> bone.This is woven in <strong>the</strong> early stages <strong>and</strong> lamellar later.8.3.1.2 CholesteatomaCholesteatoma is <strong>the</strong> presence <strong>of</strong> stratified squamousepi<strong>the</strong>lium in considerable quantities in <strong>the</strong> middle ear.The common acquired form <strong>of</strong> cholesteatoma is associatedwith severe otitis media.Stratified squamous epi<strong>the</strong>lium in <strong>the</strong> normal foetalmiddle ear: small colonies <strong>of</strong> cells being epidermoidin nature as confirmed by immunohistochemistryare found near <strong>the</strong> tympanic membrane on <strong>the</strong> lateralanterior superior surface <strong>of</strong> <strong>the</strong> middle ear in everytemporal bone after 15 weeks’ gestation. These epidermalcolonies, which are known as “epidermoid formations”,increase significantly in size with increasing age <strong>and</strong>pari passu undergo increasing epidermoid differentiation[58]. During <strong>the</strong> first postpartum year <strong>the</strong>se epidermoidformations disappear. It is possible that <strong>the</strong> entry<strong>and</strong> growth <strong>of</strong> epidermoid formations in <strong>the</strong> foetal middleear may lead to a local cellular immunity as a defencemechanism against <strong>the</strong> entry <strong>of</strong> keratinocytes into <strong>the</strong>middle ear. This could cause <strong>the</strong> eventual dissolution<strong>of</strong> all epidermoid formations. If immunity is delayed ordefective epidermis could continue to grow <strong>and</strong> lead tocongenital cholesteatoma.Stratified squamous epi<strong>the</strong>lium in <strong>the</strong> middle ear <strong>of</strong> ayoung child (congenital cholesteatoma): congenital cholesteatomais seen, in most cases as a spherical whitish


Ear <strong>and</strong> Temporal Bone Chapter 8 245object in <strong>the</strong> anterosuperior part <strong>of</strong> <strong>the</strong> tympanic cavitybehind an intact tympanic membrane (Fig. 8.8). In somecases <strong>the</strong> lesion may fill most <strong>of</strong> <strong>the</strong> tympanic cavity. Atoperation <strong>the</strong> cholesteatoma is reported usually to be acyst in <strong>the</strong> anterosuperior part <strong>of</strong> <strong>the</strong> middle ear. Boneerosion is not present when <strong>the</strong> cholesteatoma is small.In larger lesions some degree <strong>of</strong> this change is present[61] <strong>and</strong> eventually it may enlarge to involve <strong>the</strong> mastoid,cause a perforation <strong>of</strong> <strong>the</strong> tympanic membrane <strong>and</strong>even grow into <strong>the</strong> middle cranial fossa [34] so that itbecomes indistinguishable from acquired cholesteatoma(see below). Indeed, it is possible that many cases <strong>of</strong>acquired cholesteatoma originated from congenital cholesteatoma.In approximately 10% <strong>of</strong> cases <strong>the</strong> cholesteatoma isnot cystic, but open <strong>and</strong> shows layers <strong>of</strong> squames <strong>and</strong> amatrix (living basal <strong>and</strong> malpighian layers <strong>of</strong> epidermis)that is plastered to <strong>the</strong> wall <strong>of</strong> <strong>the</strong> tympanic cavity [19].The microscopic appearances <strong>of</strong> <strong>the</strong> matrix <strong>of</strong> congenitalcholesteatoma are those <strong>of</strong> skin epidermis, comprisinga single row <strong>of</strong> basal cells, several rows <strong>of</strong> malpighiancells <strong>and</strong> a thin granular layer. The surface <strong>of</strong> dead, keratinoussquames merges with <strong>the</strong> keratinous contents <strong>of</strong><strong>the</strong> cyst, or lamellae in <strong>the</strong> case <strong>of</strong> <strong>the</strong> open type. When<strong>the</strong> histological appearance <strong>of</strong> <strong>the</strong>se cases is comparedwith that <strong>of</strong> acquired cholesteatoma, little difference canbe seen.Stratified squamous epi<strong>the</strong>lium in <strong>the</strong> middle ear <strong>of</strong>an older child or adult, acquired cholesteatoma: typicallyin acquired cholesteatoma a lesion far more commonthan that <strong>of</strong> congenital cholesteatoma, <strong>the</strong> patientpresents with a foul-smelling aural discharge <strong>and</strong> conductivehearing loss. On examination <strong>of</strong> <strong>the</strong> tympanicmembrane <strong>the</strong>re is, in most cases, a perforation <strong>of</strong> <strong>the</strong>superior or posterosuperior margin. The cholesteatomaappears as a pearly grey structure in <strong>the</strong> middle ear cavity.The wall <strong>of</strong> <strong>the</strong> cyst may <strong>of</strong>ten be seen as a thin membrane.The cholesteatoma is usually situated in <strong>the</strong> upperposterior part <strong>of</strong> <strong>the</strong> middle ear cleft <strong>and</strong> dischargesusually through a perforation <strong>of</strong> <strong>the</strong> pars flaccida<strong>of</strong> <strong>the</strong> tympanic membrane, sometimes through aperforation located at <strong>the</strong> edge <strong>of</strong> <strong>the</strong> tympanic membranenear <strong>the</strong> annulus. The cholesteatoma may extendthrough <strong>the</strong> aditus into <strong>the</strong> mastoid antrum <strong>and</strong> mastoidair cells. Frequently, <strong>the</strong> outline <strong>of</strong> <strong>the</strong> cholesteatomatoussac is adapted to that <strong>of</strong> normal structuressuch as ossicles. Chronic inflammatory changes are alwayspresent. In most cases at least one ossicle is seriouslydamaged, thus interrupting <strong>the</strong> continuity <strong>of</strong> <strong>the</strong>ossicular chain. The scutum, <strong>the</strong> upper part <strong>of</strong> <strong>the</strong> bonyring <strong>of</strong> <strong>the</strong> tympanic opening, is eroded in most cholesteatomas.Under <strong>the</strong> microscope acquired cholesteatoma isusually “open” ra<strong>the</strong>r than “closed” or cystic. The pearlyFig. 8.8. Congenital cholesteatoma seen as a small cyst in <strong>the</strong> anterosuperiorpart <strong>of</strong> <strong>the</strong> middle ear. Reproduced from Michaels<strong>and</strong> Hellquist [68]Fig. 8.9. Acquired cholesteatoma showing keratinising stratifiedsquamous epi<strong>the</strong>lium with a granular layer. Reproduced from Michaels<strong>and</strong> Hellquist [68]material <strong>of</strong> <strong>the</strong> cholesteatoma consists <strong>of</strong> dead, fully differentiatedanucleate keratin squames. This is <strong>the</strong> corneallayer <strong>of</strong> <strong>the</strong> squamous cell epi<strong>the</strong>lium. As in any normalstratified epi<strong>the</strong>lium <strong>the</strong>re are one to three basal layers<strong>of</strong> cells above which is a prickle (malpighian or spinous)layer composed <strong>of</strong> five or six rows <strong>of</strong> cells withintercellular bridges (Fig. 8.9).The deeper layers <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong> cholesteatomamatrix frequently show evidence <strong>of</strong> activity in <strong>the</strong>form <strong>of</strong> downgrowths into <strong>the</strong> underlying connectivetissue (Fig. 8.10). Such excessive activity has been confirmedby:1. The strong expression <strong>of</strong> cytokeratin 16, a marker forhyperproliferative keratinocytes, by cholesteatoma,but its absence in middle ear <strong>and</strong> external ear epi<strong>the</strong>lium,except in <strong>the</strong> annulus region <strong>of</strong> <strong>the</strong> externaltympanic membrane epi<strong>the</strong>lium [13];


246 L. Michaels8Fig. 8.10. Acquired cholesteatoma showing downgrowths from<strong>the</strong> deeper layer <strong>of</strong> cholesteatoma epi<strong>the</strong>lium2. The strong expression <strong>of</strong> MIB1, an antigen related toKi-67, which also indicates hyperproliferative activity[113];3. Counts <strong>of</strong> silver-stained argyrophil nucleolar organiserregions, a technique that likewise displaysproliferative activity, showed significantly largernumbers <strong>of</strong> <strong>the</strong>se structures in <strong>the</strong> nuclei <strong>of</strong> acquiredcholesteatoma compared with those <strong>of</strong> <strong>the</strong>epidermis <strong>of</strong> <strong>the</strong> deep external auditory meatal skin[115];4. Acquired cholesteatomatous epi<strong>the</strong>lium shows anabnormally high concentration <strong>of</strong> IL-1, TGF-alpha,EGF-R <strong>and</strong> 4F2, all being growth factors [114] indicatinggreater growth <strong>and</strong> differentiating activitythan is present in normal epidermis.Congenital cholesteatoma probably arises due to <strong>the</strong>continued growth <strong>of</strong> <strong>the</strong> epidermoid formation. Thisstructure is derived from external ear epidermis (seeabove). It seems likely that acquired cholesteatoma isalso derived from entry <strong>of</strong> external ear canal epidermisinto <strong>the</strong> middle ear. This is clearly shown in those cases<strong>of</strong> acquired cholesteatoma that follow blast injury withperforation <strong>of</strong> <strong>the</strong> tympanic membrane at <strong>the</strong> time <strong>of</strong><strong>the</strong> injury [56]. Acquired cholesteatoma is also knownto follow <strong>the</strong> retraction pocket <strong>of</strong> <strong>the</strong> tympanic membrane.This is not due to obstruction <strong>of</strong> <strong>the</strong> mouth <strong>of</strong> aretraction pocket, but ra<strong>the</strong>r, it seems, to <strong>the</strong> ingrowth<strong>of</strong> a b<strong>and</strong> <strong>of</strong> stratified squamous epi<strong>the</strong>lium from <strong>the</strong>fundus <strong>of</strong> <strong>the</strong> retraction pocket deeply into <strong>the</strong> middleear (Fig. 8.11) [122]. A similar entry <strong>of</strong> stratifiedsquamous epi<strong>the</strong>lium from <strong>the</strong> external ear epidermisthrough <strong>the</strong> tympanic membrane may sometimes beobserved in human temporal bone sections in cases <strong>of</strong>severe otitis media (Fig. 8.12). The placement <strong>of</strong> irritantsor bacteria into <strong>the</strong> middle ear cavity <strong>of</strong> animalsFig. 8.11. The fundus <strong>of</strong> a retraction pocket is seen in <strong>the</strong> top righth<strong>and</strong> part <strong>of</strong> <strong>the</strong> illustration. Emanating from it <strong>and</strong> passing downto <strong>the</strong> bottom centre is a b<strong>and</strong> <strong>of</strong> stratified squamous epi<strong>the</strong>lium,within which is a bluish staining zone <strong>of</strong> stratum granulosum <strong>and</strong>keratin – a “mini-cholesteatoma”. An ossicle lies to <strong>the</strong> right <strong>of</strong> <strong>the</strong>epidermal b<strong>and</strong>. It is an eroded incus, damage to which has probablybeen caused by previous otitis mediaFig. 8.12. A thin str<strong>and</strong> <strong>of</strong> stratified squamous epi<strong>the</strong>lium is seenpassing from <strong>the</strong> epidermis <strong>of</strong> <strong>the</strong> tympanic membrane towards<strong>the</strong> middle ear epi<strong>the</strong>lium, almost touching it. To <strong>the</strong> left <strong>of</strong> thisepidermal b<strong>and</strong> <strong>the</strong> middle collagenous layer <strong>of</strong> <strong>the</strong> eardrum isdistorted by a deposit <strong>of</strong> tympanosclerosis, composed <strong>of</strong> partiallycalcified hyaline collagenhas been known to provoke an otitis media that is associatedwith epidermal invasion through <strong>the</strong> tympanicmembrane with <strong>the</strong> subsequent development <strong>of</strong> cholesteatoma.In chinchillas, destruction <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium<strong>of</strong> both middle ear <strong>and</strong> lateral tympanic membranesurfaces takes place in <strong>the</strong> early stages <strong>of</strong> such an artificialacute otitis media, induced by insertion <strong>of</strong> propyleneglycol into <strong>the</strong> middle ear. This is followed by reepi<strong>the</strong>lialisationwith hyperplastic epidermal cells <strong>and</strong><strong>the</strong>n penetration <strong>of</strong> <strong>the</strong> thickened fibrous layer <strong>of</strong> <strong>the</strong>tympanic membrane by <strong>the</strong> epidermal cells to reach<strong>the</strong> middle ear cavity <strong>and</strong> <strong>the</strong> formation <strong>of</strong> keratinousmasses in <strong>the</strong> middle ear typical <strong>of</strong> cholesteatoma [60,127].


Ear <strong>and</strong> Temporal Bone Chapter 8 2478.3.1.3 Unusual Inflammatory LesionsTuberculous otitis media is an unusual form <strong>of</strong> chronicotitis media, which is generally associated with activepulmonary tuberculosis. In <strong>the</strong> initial stages multipleperforations <strong>of</strong> <strong>the</strong> tympanic membrane develop.Granulations in <strong>the</strong> middle ear may appear pale <strong>and</strong>are <strong>of</strong>ten pr<strong>of</strong>use. Complications, especially involvement<strong>of</strong> <strong>the</strong> facial nerve, are more frequent than in <strong>the</strong>commoner form <strong>of</strong> chronic otitis media. The diagnosisis usually made by histopathological examination <strong>of</strong>biopsy material from middle ear contents. This is <strong>of</strong>tendelayed because surgeons are reluctant to take biopsiesfrom cases <strong>of</strong> chronic otitis media that seem fairly typical.Culture <strong>of</strong> middle ear inflammatory tissue may producetubercle bacilli. Histological examination showstuberculoid granulation tissue composed <strong>of</strong> epi<strong>the</strong>lioidcells, Langerhans giant cells <strong>and</strong> areas <strong>of</strong> caseation situatedin <strong>the</strong> middle ear mucosa. There is much bone destruction.Acid-fast bacilli are found with difficulty in<strong>the</strong> granulomatous material.8.3.2 Neoplasms<strong>and</strong> Lesions ResemblingNeoplasms8.3.2.1 Choristoma(Salivary Gl<strong>and</strong>, Glial<strong>and</strong> Sebaceous Types)A hamartoma is a focal overgrowth, in improper proportions,<strong>of</strong> tissues normally present in that part <strong>of</strong> <strong>the</strong>body. A choristoma is similar to hamartoma, exceptthat <strong>the</strong> tissues <strong>of</strong> which it is composed are not normallypresent in <strong>the</strong> part <strong>of</strong> <strong>the</strong> body where it is found.Choristomas are occasionally seen in <strong>the</strong> middle ear.They are composed <strong>of</strong> one or o<strong>the</strong>r <strong>of</strong> three types <strong>of</strong>tissue: salivary gl<strong>and</strong>, glial or sebaceous gl<strong>and</strong>ular tissue.Salivary gl<strong>and</strong> choristomas consist as a rule <strong>of</strong> mixedmucous <strong>and</strong> serous elements like <strong>the</strong> normal subm<strong>and</strong>ibularor sublingual gl<strong>and</strong>, but unlike <strong>the</strong> parotidgl<strong>and</strong>. The lesion typically consists <strong>of</strong> a lobulated mass<strong>of</strong> histologically normal salivary gl<strong>and</strong> tissue in <strong>the</strong> middleear attached posteriorly in <strong>the</strong> region <strong>of</strong> <strong>the</strong> oval window.Frequently, <strong>the</strong> mass is intimately associated with<strong>the</strong> facial nerve. There are usually absent or malformedossicles [44].Glial choristomas are composed largely <strong>of</strong> astrocyticcells with large amounts <strong>of</strong> glial fibrils, <strong>the</strong> identity<strong>of</strong> which may be confirmed by immunohistochemicalstaining for glial acidic fibrillary protein. When suchmasses are identified in biopsy material from <strong>the</strong> middleFig. 8.13. Adenoma <strong>of</strong> <strong>the</strong> middle ear. Reproduced from Michaels<strong>and</strong> Hellquist [68]ear a bony deficit with consequent herniation <strong>of</strong> braintissue into <strong>the</strong> middle ear should be ruled out [53]. Threecases <strong>of</strong> heterotopic brain tissue in <strong>the</strong> middle ear associatedwith cholesteatoma have been reported [62]. It ispossible that in all three, brain herniation occurred as aresult <strong>of</strong> inflammatory damage to <strong>the</strong> tegmen tympani.Spontaneous herniations <strong>of</strong> brain (encephaloceles) mayoccur into <strong>the</strong> middle ear through a congenital deficiency<strong>of</strong> <strong>the</strong> tegmen or o<strong>the</strong>r sites [47].A case <strong>of</strong> sebaceous choristoma <strong>of</strong> <strong>the</strong> middle ear hasbeen described [82].8.3.2.2 AdenomaICD-O:8140/0Adenoma is <strong>the</strong> commonest neoplasm <strong>of</strong> <strong>the</strong> middle ear.The epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong> middle ear has a propensity towardsgl<strong>and</strong> formation in otitis media (see above) <strong>and</strong>adenoma would seem to represent a benign neoplastictransformation <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium along <strong>the</strong> same lines.The neoplasm has been described as being white, yellow,grey or reddish brown at operation <strong>and</strong> unlike paragangliomais usually not vascular. It is usually situatedin <strong>the</strong> middle ear cavity, sometimes extending into <strong>the</strong>mastoid. It seems to peel away from <strong>the</strong> walls <strong>of</strong> <strong>the</strong> surroundingmiddle ear with ease, although ossicles maysometimes be entrapped in <strong>the</strong> tumour mass <strong>and</strong> mayeven show destruction.Adenoma is formed by closely apposed small gl<strong>and</strong>swith a “back to back” appearance (Fig. 8.13). In someplaces a solid or trabecular arrangement is present.Sheet-like, disorganised areas are seen in which <strong>the</strong>gl<strong>and</strong>ular pattern appears to be lost. This may be artefactual<strong>and</strong> related to <strong>the</strong> effects <strong>of</strong> trauma used intaking <strong>the</strong> biopsy specimen on <strong>the</strong> delicate structure <strong>of</strong><strong>the</strong> cells, but <strong>the</strong> appearance may erroneously lead oneto suspect malignancy. The cells are regular, cuboidalor columnar <strong>and</strong> may enclose luminal secretion. A dis-


248 L. Michaels8tinct <strong>and</strong> predominant “plasmacytoid” appearance <strong>of</strong><strong>the</strong> epi<strong>the</strong>lial cells <strong>of</strong> <strong>the</strong> neoplasm may be displayed[92].No myoepi<strong>the</strong>lial layer is seen. PAS <strong>and</strong> Alcian bluestains may be positive for mucoprotein secretion in <strong>the</strong>gl<strong>and</strong> lumina <strong>and</strong> in <strong>the</strong> cytoplasm <strong>of</strong> <strong>the</strong> tumour cells.Benign gl<strong>and</strong>ular tumours <strong>of</strong> <strong>the</strong> middle ear were notdescribed until 1976 [26, 45]. It was soon reported that agl<strong>and</strong>ular tumour <strong>of</strong> <strong>the</strong> middle ear, o<strong>the</strong>rwise apparentlyidentical to an adenoma, was Grimelius positive <strong>and</strong>on electron microscopy, showed numerous membraneboundgranules [75]. The use <strong>of</strong> immunohistochemistryfrom 1987, fur<strong>the</strong>r confirmed <strong>the</strong> presence <strong>of</strong> neuroendocrinefeatures in some <strong>of</strong> <strong>the</strong>se neoplasms [108]. In an investigation<strong>of</strong> five cases <strong>of</strong> adenoma <strong>of</strong> <strong>the</strong> middle ear bylight microscopic methods, immunohistochemistry <strong>and</strong>transmission electron microscopy, <strong>the</strong> gl<strong>and</strong>ular areas <strong>of</strong><strong>the</strong> tumour in each patient showed bidirectional mucinous<strong>and</strong> neuroendocrine differentiation. This was demonstratedby <strong>the</strong> presence <strong>of</strong> two cell types. Apically situateddark cells contained mucous granules; <strong>the</strong>se cellswere negative for neuroendocrine markers. Basally situatedcells contained neuroendocrine granules; <strong>the</strong>se cellswere positive for neuroendocrine markers – vasoactiveintestinal polypeptides or neuron-specific enolase [120].It seems likely that <strong>the</strong>re is but a single benign gl<strong>and</strong>ularneoplasm <strong>of</strong> <strong>the</strong> middle ear, <strong>the</strong> adenoma. Neuroendocrineas well as mucinous differentiation is frequent,perhaps universal, in <strong>the</strong>se neoplasms. Contraryto what has been suggested by some authors <strong>the</strong>re is noevidence that <strong>the</strong> presence <strong>of</strong> neuroendocrine differentiationreflects a more aggressive potential in adenomas,which are benign tumours.8.3.2.3 Papillary TumoursFig. 8.14. Aggressive papillary tumour <strong>of</strong> <strong>the</strong> middle earICD-O:8260/0, 8260/3Aggressive papillary tumour is characterised by a papillary,non-stratified epi<strong>the</strong>lial histological pattern thatshows aggressive, <strong>of</strong>ten invasive behaviour. Forty-sixcases in which <strong>the</strong> temporal bone was affected by thisneoplasm were collected from <strong>the</strong> literature in 1994 [31].Some <strong>of</strong> <strong>the</strong>se had been reported as low-grade adenocarcinoma<strong>of</strong> probable endolymphatic sac origin (seebelow <strong>and</strong> also Sect. 8.4.2.3) [41]. I have reviewed each<strong>of</strong> <strong>the</strong> case reports cited in <strong>the</strong>se two studies toge<strong>the</strong>rwith cases reported in <strong>the</strong> literature more recently, <strong>and</strong>this has produced a total <strong>of</strong> 25 cases in which <strong>the</strong> middleear was definitely involved in <strong>the</strong> neoplasm. Some <strong>of</strong> <strong>the</strong>literature sources reported more than one case [1, 9, 10,21, 31–33, 35, 38, 41, 50, 86, 88, 109, 111, 118].The 25 literature cases with this middle ear neoplasmcomprised 18 females <strong>and</strong> 7 males. The age-range at time<strong>of</strong> diagnosis was between 16 <strong>and</strong> 55 years with a medianage <strong>of</strong> 33 <strong>and</strong> a mean age <strong>of</strong> 34 years. In many <strong>of</strong> <strong>the</strong> cases,however, <strong>the</strong> patient had already suffered symptomssubsequently ascribable to <strong>the</strong> tumour for some yearswhen <strong>the</strong> diagnosis was made, so that <strong>the</strong> age <strong>of</strong> onsetmay be considerably younger than is suggested.The tumour is found in any area <strong>of</strong> <strong>the</strong> middle ear,including <strong>the</strong> mastoid process <strong>and</strong> air cells <strong>and</strong> may fill<strong>the</strong> tympanic cavity. In all <strong>of</strong> <strong>the</strong> described cases, exceptthree [21, 109, 118], <strong>the</strong>re was extensive invasion outside<strong>the</strong> middle ear, involving <strong>the</strong> apical portion <strong>of</strong> <strong>the</strong> petrousbone in most <strong>and</strong> in a few <strong>the</strong> tumour reached <strong>the</strong>cerebellopontine angle <strong>and</strong> <strong>the</strong> cerebellum.It has been suggested that cases <strong>of</strong> aggressive papillarymiddle ear tumour with widespread involvement <strong>of</strong><strong>the</strong> temporal bone may arise from a primary papillaryadenocarcinoma <strong>of</strong> <strong>the</strong> endolymphatic sac (endolymphaticsac tumour, low-grade adenocarcinoma <strong>of</strong> probableendolymphatic sac origin) [41]. The frequent association<strong>of</strong> papillary tumours in <strong>the</strong> middle ear with apicalpetrous bone neoplasia <strong>of</strong> <strong>the</strong> same type, <strong>the</strong> similarity<strong>of</strong> <strong>the</strong> histological appearances <strong>of</strong> <strong>the</strong> neoplasmin <strong>the</strong> two regions <strong>and</strong> <strong>the</strong> association <strong>of</strong> some cases <strong>of</strong>papillary tumours in both regions with von Hippel-Lindaudisease would seem to favour this concept. Such anorigin has not yet been confirmed by autopsy study. Indeed,in <strong>the</strong> single description <strong>of</strong> <strong>the</strong> pathological changes<strong>of</strong> aggressive papillary tumour <strong>of</strong> <strong>the</strong> middle ear in anautopsy-acquired temporal bone, widespread deposits<strong>of</strong> tumour at inner ear sites are depicted, but no mentionis made <strong>of</strong> involvement <strong>of</strong> <strong>the</strong> endolymphatic sacor duct [100]. Thus, a middle ear origin for some cases<strong>of</strong> this neoplasm at least has not been definitely excluded.Whatever <strong>the</strong> site or sites <strong>of</strong> origin <strong>of</strong> this tumour, itshould be recognised that papillary epi<strong>the</strong>lial tumour <strong>of</strong><strong>the</strong> middle ear is an aggressive neoplasm, in contrast to<strong>the</strong> non-papillary adenoma <strong>of</strong> <strong>the</strong> middle ear, which isquite benign [73].In view <strong>of</strong> <strong>the</strong> association <strong>of</strong> some cases <strong>of</strong> von Hippel-Lindaudisease with aggressive papillary middle eartumours it is suggested that <strong>the</strong> clinical assessment <strong>of</strong>


Ear <strong>and</strong> Temporal Bone Chapter 8 249each case with <strong>the</strong> latter neoplasm should include an investigation<strong>of</strong> <strong>the</strong> gene mutations <strong>of</strong> von Hippel-Lindaudisease.In most cases <strong>of</strong> this neoplasm, clinical <strong>and</strong> audiologicalfeatures point to a middle ear lesion. Suspicion<strong>of</strong> a neoplasm <strong>of</strong> <strong>the</strong> middle ear is enhanced by <strong>the</strong> otoscopicfeatures.The middle ear cleft, including <strong>the</strong> mastoid air cells,is usually filled with <strong>the</strong> papillary tumour. Bone invasionis <strong>of</strong>ten seen. A papillary gl<strong>and</strong>ular pattern is presentwith complex interdigitating papillae lying looselyor infiltrating fibrous connective tissue. The papillae arelined with a single layer <strong>of</strong> low cuboidal to columnar epi<strong>the</strong>lialcells with uniform nuclei, eosinophilic cytoplasm<strong>and</strong> indistinct cell borders (Fig. 8.14). Thyroid folliclelikeareas may be present similar to those seen in endolymphaticsac carcinoma (see below).Markers for cytokeratin, epi<strong>the</strong>lial membrane antigen<strong>and</strong> S-100 are positive. The absence <strong>of</strong> thyroglobulinmust be determined to exclude metastatic papillarycarcinoma <strong>of</strong> <strong>the</strong> thyroid. Markers for CK 7, CK 20 <strong>and</strong>carcinoembryonic antigen may also be useful to excludemetastatic deposits from lung <strong>and</strong> colon.Schneiderian papillomas (ICD-O:8121/0) are tumours<strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasal sinuses that are statedto be derived from <strong>the</strong> Schneiderian epi<strong>the</strong>lium, a termused to denote <strong>the</strong> normal respiratory-type ciliated epi<strong>the</strong>lium<strong>of</strong> <strong>the</strong> nose <strong>and</strong> paranasal sinuses. Three types<strong>of</strong> such papillomas are described: inverted (endophytic),exophytic (fungiform, everted) <strong>and</strong> oncocytic (cylindriccell). Intermediate types are said to be found among<strong>the</strong> three forms [73]. It has, however, been denied thatsuch intermediate forms exist <strong>and</strong> it is suggested that <strong>the</strong>three types are each separate <strong>and</strong> distinct entities <strong>of</strong> <strong>the</strong>nose <strong>and</strong> paranasal sinuses [71]. Of <strong>the</strong> three histologicalforms only inverted papilloma is characteristcally asinonasal neoplasm. The o<strong>the</strong>r two types <strong>of</strong> Schneiderian-typepapilloma may be seen at o<strong>the</strong>r sites. Low-gradesquamous carcinoma in <strong>the</strong> nose may sometimes bemistaken for inverted papilloma [67].Fourteen cases <strong>of</strong> middle ear tumours purportedlyresembling Schneiderian-type papilloma have beenfound in <strong>the</strong> literature. Each <strong>of</strong> <strong>the</strong>se cases is listed inTable 8.1; wherever possible <strong>the</strong> histological appearancesare summarised in <strong>the</strong> table. In some insufficient orno histological description was given. In two cases onlywere <strong>the</strong> features <strong>of</strong> inverted papilloma depicted: inCase 1 <strong>the</strong> term “inverted” <strong>and</strong> in Case 2 <strong>the</strong> term “endophytic”were used to describe <strong>the</strong> neoplasm. “Inverted”or “endophytic” features comprised only a portion<strong>of</strong> <strong>the</strong> tumours in <strong>the</strong> two cases. In Case 2 [52] <strong>the</strong> termtransitional cell papilloma was used. This is a term thathas been frequently applied to describe everted squamouscell papilloma. In this case inverted papillomawas found in <strong>the</strong> nasal cavity <strong>and</strong> it was suggested that<strong>the</strong> papillomas may have spread from <strong>the</strong>re to <strong>the</strong> middleear by way <strong>of</strong> <strong>the</strong> Eustachian tube. In Cases 1, 4 <strong>and</strong>10 inverted papilloma was found in <strong>the</strong> middle ear concomitantlywith in situ or invasive squamous carcinoma<strong>and</strong> it seems possible that <strong>the</strong> inverted papilloma areasmight have been, in reality, areas <strong>of</strong> low-grade squamouscarcinoma.I would suggest that a good case has not been madefor <strong>the</strong> occurrence <strong>of</strong> inverted papilloma in <strong>the</strong> middleear. Some <strong>of</strong> <strong>the</strong> lesions may have been papillomas <strong>of</strong><strong>the</strong> middle ear as described above. In Case 2 an invertedpapilloma could conceivably have colonised <strong>the</strong> middleear from <strong>the</strong> nasal cavity. Fur<strong>the</strong>r detailed descriptions<strong>of</strong> <strong>the</strong> entity are required to justify <strong>the</strong> diagnosis <strong>of</strong> sucha diagnostic category in this situation.8.3.2.4 JugulotympanicParagangliomaICD-O:8690/1Most jugulotympanic paragangliomas arise from <strong>the</strong>paraganglion situated in <strong>the</strong> wall <strong>of</strong> <strong>the</strong> jugular bulb.These tumours have been referred to as jugular paragangliomasor glomus jugulare tumours. A minorityarise from <strong>the</strong> paraganglion situated near <strong>the</strong> middleear surface <strong>of</strong> <strong>the</strong> promontory. These tumours have beenreferred to as tympanic paragangliomas or glomus tympanicumtumours. The distinction between jugular <strong>and</strong>tympanic paragangliomas can easily be made in <strong>the</strong> patientby modern imaging methods by which <strong>the</strong> jugularneoplasm is identified as arising from <strong>the</strong> jugular bulbregion <strong>and</strong> shows evidence <strong>of</strong> invasion <strong>of</strong> <strong>the</strong> petrousbone, while <strong>the</strong> tympanic neoplasm is confined to <strong>the</strong>middle ear.The gross <strong>and</strong> histological appearances <strong>of</strong> <strong>the</strong> twotypes <strong>of</strong> neoplasm in <strong>the</strong> middle ear are, however, identical.Solitary jugulotympanic paragangliomas arisepredominantly in females. The neoplasm has beenseen at ages between 13 <strong>and</strong> 85 years with a mean age <strong>of</strong>about 50 years. Most patients present with conductivehearing loss. Pain in <strong>the</strong> ear, facial palsy, haemorrhage<strong>and</strong> tinnitus are also described as symptoms <strong>of</strong> this lesion.On examination a red vascular mass is seen ei<strong>the</strong>rbehind <strong>the</strong> intact tympanic membrane or sproutingthrough <strong>the</strong> latter into <strong>the</strong> external canal. Surgicalapproach to <strong>the</strong> mass at biopsy <strong>of</strong>ten results in severebleeding.Jugulotympanic paragangliomas may also be multicentricor coexist with tumours <strong>of</strong> o<strong>the</strong>r types. Theymay be bilateral in <strong>the</strong> same patient <strong>and</strong> coexist withcarotid body paragangliomas that may be bilateral[84]. They may also coexist with adrenal gl<strong>and</strong> pheochromocytomas,which can produce hypertension. Afamilial tendency to grow paragangliomas has beennoted particularly in cases with multiple tumours <strong>of</strong>this type. In families containing patients with head


250 L. MichaelsTable 8.1. Schneiderian-type or inverted papillomas <strong>of</strong> <strong>the</strong> middle ear described in <strong>the</strong> literatureCasenumberLiteraturesourceHistological description givenPossible alternative diagnosis81 [110] “Inverted papilloma” <strong>and</strong> high-grade carcinoma Squamous cell carcinoma <strong>of</strong><strong>the</strong> middle ear2 [52] “Transitional cell papilloma”Inverted papilloma in noseInverted papilloma derivedfrom a nasal tumour3 [93] Entirely papillary Papilloma <strong>of</strong> <strong>the</strong> middle ear4 [98] Malignant change in inverted papilloma Squamous cell carcinoma<strong>of</strong> <strong>the</strong> middle ear5 [124]Case 16 [124]Case 27 [124]Case 38 [124]Case 49 [124]Case 5“Epidermoid papilloma” with “inverted” <strong>and</strong> “cylindriccell papilloma”“Epidermoid papilloma” with exophytic <strong>and</strong> endophyticgrowth“Epidermoid papilloma” with features <strong>of</strong> “cylindric cellpapilloma”“Epidermoid papilloma” with features <strong>of</strong> “cylindric cellpapilloma”“Epidermoid papilloma” with features <strong>of</strong> “cylindric cellpapilloma”Papilloma <strong>of</strong> <strong>the</strong> middle earPapilloma <strong>of</strong> middle earPapilloma <strong>of</strong> <strong>the</strong> middle earPapilloma <strong>of</strong> <strong>the</strong> middle earPapilloma <strong>of</strong> <strong>the</strong> middle ear10 [51] Squamous epi<strong>the</strong>lium with areas <strong>of</strong> carcinoma in situ Squamous carcinoma <strong>of</strong><strong>the</strong> middle ear11 [17] Papillary with areas <strong>of</strong> squamous epi<strong>the</strong>lium adjacent to Papilloma <strong>of</strong> <strong>the</strong> middle earrespiratory epi<strong>the</strong>lium12 [90]None ?Case 113 [90]?Case 214 [11]Two casesInsufficient description ?<strong>and</strong> neck, including jugulotympanic, paragangliomas<strong>the</strong>re is, unlike <strong>the</strong> solitary jugulotympanic paraganglioma,a preponderance for <strong>the</strong> male sex <strong>and</strong> inheritanceis autosomal dominant, with increased penetrancewith age [5]. There is evidence from moleculargenetic studies that <strong>the</strong> gene underlying familial paragangliomasis located on chromosome 11q proximal to<strong>the</strong> tyrosinase gene locus [59].The neoplasm is a reddish sprouting mass at its externalcanal surface. In <strong>the</strong> jugular variety <strong>the</strong> petroustemporal bone is largely replaced by red, firm material<strong>and</strong> <strong>the</strong> middle ear space is occupied by s<strong>of</strong>t neoplasm asfar as <strong>the</strong> tympanic membrane. The otic capsule is rarelyinvaded by paraganglioma. Investigation <strong>of</strong> a paragangliomain an autopsy temporal bone by <strong>the</strong> microslicingmethod showed <strong>the</strong> origin <strong>of</strong> <strong>the</strong> tumour to be in<strong>the</strong> jugular bulb region <strong>and</strong> its spread through <strong>the</strong> petrousbone <strong>and</strong> middle ear to <strong>the</strong> tympanic membrane(Fig. 8.15). The histology <strong>of</strong> paraganglioma is describedin Chap. 9 (Fig. 8.16) [55].The incidence <strong>of</strong> clinically functioning paragangliomawith symptoms <strong>and</strong> signs <strong>of</strong> norepinephrine excess,particularly hypertension, is only 1–3% [97].Jugulotympanic paraganglioma is a neoplasm <strong>of</strong> slowgrowth. The jugular variety infiltrates <strong>the</strong> petrous bone,but distant metastasis is rare. Radiation <strong>the</strong>rapy, <strong>and</strong> insome cases surgery, <strong>of</strong>fers a high rate <strong>of</strong> cure for <strong>the</strong>seneoplasms <strong>and</strong> <strong>the</strong> number <strong>of</strong> patients who do badly aftersuch <strong>the</strong>rapy is very small.8.3.2.5 Squamous Cell CarcinomaICD-O:8070/3Squamous cell carcinoma is uncommon in <strong>the</strong> middleear. It sometimes accompanies squamous cell carcinoma<strong>of</strong> <strong>the</strong> external canal or may arise solely from <strong>the</strong>middle ear epi<strong>the</strong>lium. The patient always has an auraldischarge <strong>and</strong> conductive hearing loss. Pain in <strong>the</strong> ear,bleeding <strong>and</strong> facial palsy are common.


Ear <strong>and</strong> Temporal Bone Chapter 8 251Fig. 8.15. Microsliced specimen <strong>of</strong> jugular paraganglioma removedat autopsy. Two slices <strong>of</strong> <strong>the</strong> temporal bone in <strong>the</strong> region<strong>of</strong> <strong>the</strong> neoplasm are seen. The one on <strong>the</strong> left shows invasion <strong>of</strong><strong>the</strong> temporal bone by <strong>the</strong> reddish paraganglioma from its apicalregion as far as <strong>the</strong> tympanic membrane. The slice on <strong>the</strong> right istaken at a higher level <strong>and</strong> shows sparing <strong>of</strong> <strong>the</strong> cochlea <strong>and</strong> bonylabyrinth by <strong>the</strong> tumourIn microscopic sections <strong>the</strong> tumour may be seen arisingfrom surface stratified squamous epi<strong>the</strong>lium, itselfmetaplastic from <strong>the</strong> normal cubical epi<strong>the</strong>lium.In certain areas an origin directly from basal layers <strong>of</strong>cubical or columnar epi<strong>the</strong>lium may be seen. There isno evidence that <strong>the</strong> epidermoid formation, a cell restthat occurs normally in <strong>the</strong> middle ear during development(see above), may be a source <strong>of</strong> squamous cell carcinoma.The neoplasm is squamous cell carcinoma withvariable degrees <strong>of</strong> differentiation. Atypical change <strong>and</strong>even carcinoma in situ may be seen in some parts <strong>of</strong> <strong>the</strong>middle ear epi<strong>the</strong>lium adjacent to <strong>the</strong> growth. The mode<strong>of</strong> spread <strong>of</strong> <strong>the</strong> neoplasm from <strong>the</strong> middle ear epi<strong>the</strong>liumhas been ascertained in temporal bone autopsy sections[70] <strong>and</strong> this pattern has been confirmed by imagingin living patients. The carcinoma tends to growinto <strong>and</strong> erode <strong>the</strong> thin bony plate that separates <strong>the</strong> medialwall <strong>of</strong> <strong>the</strong> middle ear, at its junction with <strong>the</strong> Eustachiantube, from <strong>the</strong> carotid canal. This bony wall isnormally up to 1 mm in thickness <strong>and</strong> may be recognisedradiologically. Having reached <strong>the</strong> carotid canal<strong>the</strong> growth will extend rapidly along <strong>the</strong> sympa<strong>the</strong>ticnerves <strong>and</strong> <strong>the</strong> tumour is <strong>the</strong>n impossible to eradicatesurgically. Ano<strong>the</strong>r important method <strong>of</strong> spread isthrough <strong>the</strong> bony walls <strong>of</strong> <strong>the</strong> posterior mastoid air cellsto <strong>the</strong> dura <strong>of</strong> <strong>the</strong> posterior surface <strong>of</strong> <strong>the</strong> temporal bone.From <strong>the</strong>re it spreads medially, enters <strong>the</strong> internal auditorymeatus <strong>and</strong> may <strong>the</strong>n invade <strong>the</strong> cochlea <strong>and</strong> vestibule.Spread into <strong>the</strong> lamellar bone in both <strong>of</strong> <strong>the</strong>se situationsis along vascular channels between bone trabeculae.A similar type <strong>of</strong> bone invasion may also occur fromo<strong>the</strong>r parts <strong>of</strong> <strong>the</strong> middle ear surface such as in <strong>the</strong> region<strong>of</strong> <strong>the</strong> facial nerve. The special bone <strong>of</strong> <strong>the</strong> otic capsuleis, on <strong>the</strong> o<strong>the</strong>r h<strong>and</strong>, peculiarly resistant to directspread <strong>of</strong> growth from tumours within <strong>the</strong> middle ear,<strong>and</strong> even <strong>the</strong> round window membrane is not invaded.When invasion does occur it takes place after entry <strong>of</strong><strong>the</strong> tumour into <strong>the</strong> internal auditory meatus <strong>and</strong> penetration<strong>of</strong> <strong>the</strong> bone by way <strong>of</strong> <strong>the</strong> filaments <strong>of</strong> <strong>the</strong> vestibular<strong>and</strong> cochlear divisions <strong>of</strong> <strong>the</strong> eighth nerve. In <strong>the</strong>later stages tumour grows extensively in <strong>the</strong> middle cranialfossa; it may also invade <strong>the</strong> condyle <strong>of</strong> <strong>the</strong> m<strong>and</strong>ible.Death is usually due to direct intracranial extension.Lymph node metastasis is unusual <strong>and</strong> spread by<strong>the</strong> bloodstream even more so [70].8.3.2.6 MeningiomaFig. 8.16. Jugular paraganglioma. The cells form small clusters,each surrounded by a row <strong>of</strong> flattened cells, probably sustentacularcells, <strong>and</strong> separated by blood vesselsICD-O:9530/0Meningioma is a benign tumour that usually growsintracerebrally, but is sometimes seen involving bonystructures around <strong>the</strong> brain including <strong>the</strong> middle ear.It arises from <strong>the</strong> pia-arachnoid cells <strong>of</strong> <strong>the</strong> meninges.These structures may be formed at a number <strong>of</strong> sites in<strong>the</strong> temporal bone, including <strong>the</strong> internal auditory meatus,<strong>the</strong> jugular foramen, <strong>the</strong> geniculate ganglion region<strong>and</strong> <strong>the</strong> ro<strong>of</strong> <strong>of</strong> <strong>the</strong> Eustachian tube. Thus, meningiomasthat arise from <strong>the</strong>m may be found in a wide area within<strong>the</strong> temporal bone itself [78].Meningioma <strong>of</strong> <strong>the</strong> middle ear affects females morethan males, shows an age range <strong>of</strong> between 10 <strong>and</strong>80 years (with a mean age <strong>of</strong> 49.6 years), with female patientspresenting at an older age (mean 52.0 years) thanmale patients (mean, 44.8 years) [116].The commonest temporal bone site for primary meningiomais in <strong>the</strong> middle ear cleft. In a recent study <strong>of</strong>36 cases, most <strong>of</strong> which involved <strong>the</strong> middle ear, but afew involved adjacent structures such as <strong>the</strong> external canalor temporal bone, only 2 showed a CNS connectionon radiography [116].Patients present clinically with hearing change, otitismedia, pain, <strong>and</strong>/or dizziness/vertigo.


252 L. Michaels8Gross appearances are those <strong>of</strong> a granular or even grittymass. Microscopically, <strong>the</strong> neoplasm takes <strong>the</strong> sameforms as any <strong>of</strong> <strong>the</strong> well-described intracranial types <strong>of</strong>meningioma. The commonest variety seen in <strong>the</strong> middleear is <strong>the</strong> meningo<strong>the</strong>lial type, in which <strong>the</strong> tumourcells form masses <strong>of</strong> epi<strong>the</strong>lioid, regular cells <strong>of</strong>ten disposedinto whorls, which may be large or small. Fibroblastic<strong>and</strong> psammomatous varieties are also sometimesseen in <strong>the</strong> middle ear.Histological diagnosis <strong>of</strong> meningioma may be difficultbecause <strong>the</strong> above features are indistinct. Under<strong>the</strong>se circumstances immunocytochemistry may be <strong>of</strong>some diagnostic value. Meningiomas are negative formost markers, including cytokeratins. The majority <strong>of</strong>meningiomas, however, are positive for vimentin <strong>and</strong>epi<strong>the</strong>lial membrane antigen.Nager’s review <strong>of</strong> temporal bone meningiomas indicatedthat only 2 out <strong>of</strong> 30 patients survived a 5-year period[78]. More recent experience <strong>of</strong> middle ear meningiomassignals a better outlook after careful local excision.In a recent study <strong>of</strong> 35 patients with follow up inwhich <strong>the</strong> tumour was sited mainly in <strong>the</strong> middle ear[116] surgical excision was used in all patients. Ten patientsdeveloped a recurrence from 5 months to 2 yearslater <strong>and</strong> 5 patients died with recurrent disease (mean,3.5 years); <strong>the</strong> remaining 30 patients were alive (n=25,mean: 19.0 years) or had died (n=5, mean: 9.5 years) <strong>of</strong>unrelated causes without evidence <strong>of</strong> disease. Meningiomas<strong>of</strong> <strong>the</strong> middle ear behave as slow-growing neoplasmswith a good overall prognosis (raw 5-year survival,83%). Extent <strong>of</strong> surgical excision is probably <strong>the</strong> mostimportant factor in determining outlook because recurrencesdevelop in 28% <strong>of</strong> cases.8.3.2.7 RhabdomyosarcomaICD-O:8900/3Rhabdomyosarcoma is seen occasionally in <strong>the</strong> middleear <strong>of</strong> young children [126]. On rare occasions it isfound in <strong>the</strong> middle ear <strong>of</strong> adults [81]. The tympanicmembrane is usually eroded by <strong>the</strong> growth, which extendsinto <strong>the</strong> external canal. Grossly, <strong>the</strong> tumour islobulated <strong>and</strong> dark red with a haemorrhagic cut surface.Almost all temporal bone rhabdomyosarcomasare <strong>of</strong> <strong>the</strong> embryonal type, displaying mainly spindleor round primitive skeletal muscle cells, some <strong>of</strong> whichhave clear cytoplasm staining positively for glycogen<strong>and</strong> o<strong>the</strong>rs have eosinophilic areas in <strong>the</strong> cytoplasm.Cross striations are unusual in this neoplasm. Immunohistochemicalmarkers for desmin, muscle-specificactin <strong>and</strong> antibodies against MyoD1 <strong>and</strong> myogeninconfirm this diagnosis.Rhabdomyosarcoma <strong>of</strong> <strong>the</strong> temporal bone is highlymalignant <strong>and</strong> spreads extensively into <strong>the</strong> cranial cavity,externally or to <strong>the</strong> pharyngeal region. Lymph node<strong>and</strong> bloodstream metastases frequently develop in <strong>the</strong>sepatients.8.3.2.8 Metastatic CarcinomaMetastasis <strong>of</strong> malignant neoplasms to <strong>the</strong> temporal boneincluding <strong>the</strong> middle ear is not uncommon. The breast is<strong>the</strong> commonest primary source <strong>of</strong> metastatic tumours,followed by lung, kidney, stomach, larynx <strong>and</strong> cutaneousmalignant melanoma [43, 48]. Two distinct modes<strong>of</strong> spread may be involved in bringing <strong>the</strong> neoplasmsfrom <strong>the</strong>ir primary sites to <strong>the</strong> middle ear: (a) alongvascular channels in <strong>the</strong> petrous bone (<strong>the</strong>se convey tumourdeposits to <strong>the</strong> temporal bone from distant sites),<strong>and</strong> (b) along nerves emanating from <strong>the</strong> internal auditorymeatus into <strong>the</strong> labyrinthine structures <strong>and</strong> bone.In this way, tumours reaching <strong>the</strong> meninges may spreadinto <strong>the</strong> temporal bone. In addition, direct spread maybring tumours into <strong>the</strong> ear from primary sites in areasadjacent to <strong>the</strong> temporal bone.8.4 Inner Ear8.4.1 Bony Labyrinth8.4.1.1 OtosclerosisOtosclerosis is a disease <strong>of</strong> <strong>the</strong> bony labyrinth, which,by involvement <strong>and</strong> fixation <strong>of</strong> <strong>the</strong> stapes footplate,leads to severe conductive hearing loss. Otosclerosishas some features <strong>of</strong> a hereditary disease, but its geneticsstill remain incompletely elucidated. Ultrastructural<strong>and</strong> immunohistochemical evidence for measles virus<strong>and</strong> isolation <strong>and</strong> identification <strong>of</strong> DNA <strong>and</strong> RNA sequences<strong>of</strong> that virus have been found in otosclerotictissue [18].Otosclerosis usually affects both ears symmetrically.The disease process is probably confined to <strong>the</strong> temporalbone. The pink swelling <strong>of</strong> otosclerosis may sometimeseven be detected clinically through a particularlytransparent tympanic membrane as a well-demarcated<strong>and</strong> pink focus near <strong>the</strong> promontory. A characteristictranslucency <strong>of</strong> bone adjacent to <strong>the</strong> cochlea <strong>and</strong> anteriorto <strong>the</strong> footplate is identified on a CT scan.The lesion always commences in <strong>the</strong> otic capsuletissue anterior to <strong>the</strong> footplate <strong>of</strong> <strong>the</strong> stapes. In thisposition it does not produce symptoms. These occurwhen <strong>the</strong> otosclerosis invades <strong>the</strong> adjacent stapes footplate<strong>and</strong> produces fixation <strong>of</strong> that structure <strong>and</strong> thusconductive hearing loss. It later spreads widely in <strong>the</strong>otic capsule <strong>and</strong> may involve <strong>the</strong> round window ligament.Blood vessels are prominent <strong>and</strong> evenly dis-


Ear <strong>and</strong> Temporal Bone Chapter 8 253Fig. 8.18. Focus <strong>of</strong> otosclerosis involving both <strong>the</strong> anterior (upper)<strong>and</strong> posterior (lower) part <strong>of</strong> footplate <strong>of</strong> <strong>the</strong> stapes. The anteriorfocus has invaded onto <strong>the</strong> footplate <strong>and</strong> <strong>the</strong> anterior crus.This would have produced fixation <strong>of</strong> <strong>the</strong> stapes <strong>and</strong> its attendantconductive hearing loss. Notice that <strong>the</strong> otosclerotic foci are moredarkly staining <strong>and</strong> vascular than <strong>the</strong> adjacent normal bone. Reproducedfrom Michaels <strong>and</strong> Hellquist [68]Fig. 8.17. Radiograph <strong>of</strong> microslice <strong>of</strong> autopsy temporal bonewith focus <strong>of</strong> otosclerosis. The focus is an area <strong>of</strong> mottled translucencyin <strong>the</strong> region <strong>of</strong> <strong>the</strong> fissula ante fenestram (arrowheads). Reproducedfrom Michaels <strong>and</strong> Hellquist [68]tributed. X-rays <strong>of</strong> temporal bone specimens show <strong>the</strong>well-defined lesion as a patch <strong>of</strong> mottled translucency(Fig. 8.17).The histological characteristic <strong>of</strong> otosclerosis is <strong>the</strong>presence <strong>of</strong> trabeculae <strong>of</strong> new bone, mostly <strong>of</strong> <strong>the</strong> woventype with marked vascularity. This contrasts with<strong>the</strong> well-developed lamellar bone under <strong>the</strong> outer periosteum,<strong>the</strong> endochondral middle layer <strong>and</strong> <strong>the</strong> endosteallayer <strong>of</strong> <strong>the</strong> otic capsule, a sharply demarcated edgebetween normal <strong>and</strong> otosclerotic bone being a prominentfeature. In most places osteocytes are very abundantwithin <strong>the</strong> woven bone. The footplate <strong>of</strong> <strong>the</strong> stapesis <strong>of</strong>ten invaded by otosclerotic bone, <strong>and</strong> <strong>the</strong> lowerend <strong>of</strong> <strong>the</strong> anterior crus <strong>of</strong> <strong>the</strong> stapes is sometimes invaded(Fig. 8.18). Otosclerotic bone sometimes reaches<strong>the</strong> endosteum <strong>of</strong> <strong>the</strong> cochlear capsule. In some casesit may lead to a fibrous reaction deep to <strong>the</strong> spiral ligament.These changes are probably <strong>the</strong> basis <strong>of</strong> <strong>the</strong> sensorineuralhearing loss that is also occasionally found incases <strong>of</strong> otosclerosis.Stapedectomy with insertion <strong>of</strong> a pros<strong>the</strong>sis to replace<strong>the</strong> fixed stapes <strong>and</strong> so reinstate <strong>the</strong> mobility <strong>of</strong> <strong>the</strong>ossicular chain is frequently performed as a treatmentfor otosclerosis. If <strong>the</strong> specimen <strong>of</strong> stapes is composed<strong>of</strong> <strong>the</strong> head <strong>and</strong> crura only microscopical examinationwill not show <strong>the</strong> changes <strong>of</strong> otosclerosis. On <strong>the</strong> o<strong>the</strong>rh<strong>and</strong>, if <strong>the</strong> whole stapes is removed, as is usually <strong>the</strong>case, otosclerotic bone will possibly be observed in sections<strong>of</strong> <strong>the</strong> anterior part <strong>of</strong> <strong>the</strong> footplate.8.4.1.2 Paget’s DiseasePaget’s disease (osteitis deformans) is a common conditionaffecting particularly <strong>the</strong> skull, pelvis, vertebralcolumn <strong>and</strong> femur in people over 40 years <strong>of</strong> age. Thecause is not yet certain, but <strong>the</strong> presence in many cases<strong>of</strong> paramyxovirus-like structures seen within osteoclastshas prompted <strong>the</strong> suggestion that Paget’s diseasemay be <strong>of</strong> viral aetiology <strong>and</strong> <strong>the</strong> measles virus <strong>and</strong>canine distemper viruses have been under scrutiny asc<strong>and</strong>idates. The pathological change is one <strong>of</strong> activebone formation proceeding alongside active bone destruction.The affected bones are enlarged, porous <strong>and</strong>deformed. Microscopically, bone formation is seen intrabeculae <strong>of</strong> bone with a lining <strong>of</strong> numerous osteoblasts.A mosaic appearance is formed by <strong>the</strong> frequentsuccessive deposition <strong>of</strong> bone, cessation <strong>of</strong> depositionresulting in thin, blue “cement lines”, followed againby resumption <strong>of</strong> deposition <strong>and</strong> its cessation, <strong>and</strong> soproduction <strong>of</strong> fur<strong>the</strong>r cement lines. Bone destructionis shown by <strong>the</strong> presence <strong>of</strong> numerous, large osteoclasticgiant cells with Howship’s lacunae. Areas <strong>of</strong> chronicinflammatory exudate intermixed with <strong>the</strong> bone arecommon.In <strong>the</strong> temporal bone <strong>the</strong> petrous apex, <strong>the</strong> mastoid<strong>and</strong> <strong>the</strong> bony part <strong>of</strong> <strong>the</strong> Eustachian tube are most frequentlyaffected [23, 79]. The periosteal part <strong>of</strong> <strong>the</strong> bonylabyrinth is <strong>the</strong> first to undergo pagetoid changes <strong>and</strong><strong>the</strong> pagetoid changes spread through <strong>the</strong> bone towards<strong>the</strong> membranous labyrinth, usually with a sharp line <strong>of</strong>


254 L. Michaelsdemarcation between <strong>the</strong> pagetoid area <strong>and</strong> <strong>the</strong> normalbony labyrinth.8.4.1.3 Osteogenesis Imperfecta8Osteogenesis imperfecta is a general bone disease with atriad <strong>of</strong> clinical features: multiple fractures, blue sclerae<strong>and</strong> conductive hearing loss. There is a congenital recessiveform in newborns that is <strong>of</strong>ten rapidly fatal <strong>and</strong>a tardive one in adults that is inherited as a mendeli<strong>and</strong>ominant <strong>and</strong> is more benign. Mutations <strong>of</strong> type I collagengenes have been established as <strong>the</strong> underlying causeleading to a general disturbance in <strong>the</strong> development <strong>of</strong>collagen, hence <strong>the</strong> thin sclerae appearing blue as well aspoorly formed bone tissue.In <strong>the</strong> long bones <strong>the</strong> resorption <strong>of</strong> cartilage in <strong>the</strong>development <strong>of</strong> bone is normal, but <strong>the</strong> bony trabeculae<strong>the</strong>mselves are poorly formed <strong>and</strong> <strong>the</strong> same may beseen in <strong>the</strong> temporal bone [46]. The ossicles in <strong>the</strong> tardiveform are very thin <strong>and</strong> subject to fractures. The stapesfootplate is also frequently fixed. The disturbance inlamellar bone formation can lead to extreme thinness,dehiscence, <strong>and</strong> non-union <strong>of</strong> <strong>the</strong> stapedial superstructurewith <strong>the</strong> footplate, or thickening with fixation <strong>of</strong><strong>the</strong> footplate. The nature <strong>of</strong> <strong>the</strong> bony tissue causing thisfixation is problematical. It has been suggested that osteogenesisimperfecta can be associated with otosclerosisso that <strong>the</strong> fixation is indeed otosclerotic [28]. Otosclerosis,like osteogenesis imperfecta, may indeed bepart <strong>of</strong> a general connective tissue disturbance [4].Indeed, some cases <strong>of</strong> clinical otosclerosis may berelated to mutations within <strong>the</strong> COL1A1 gene that aresimilar to those found in mild forms <strong>of</strong> osteogenesis imperfecta[63].8.4.1.4 OsteopetrosisFig. 8.19. Cytomegalovirus infection <strong>of</strong> vestibular nerve in <strong>the</strong>region <strong>of</strong> Scarpa’s ganglion. The nerve is inflamed with <strong>the</strong> infiltration<strong>of</strong> neutrophils, leucocytes <strong>and</strong> plasma cells. Four enlargedcells, three to <strong>the</strong> left <strong>of</strong> centre, <strong>the</strong> fourth on <strong>the</strong> far right) withpurplish inclusions, each surrounded by a pale halo, features characteristic<strong>of</strong> cytomegalovirus infection, can be identified. Autopsyfindings in a patient with AIDS. Reproduced from Michaels <strong>and</strong>Hellquist [68]Osteopetrosis (<strong>of</strong>ten known as marble bone disease)is a rare disease <strong>of</strong> bone, in which <strong>the</strong>re is a failure toabsorb calcified cartilage <strong>and</strong> primitive bone due todeficient activity <strong>of</strong> osteoclasts. A relatively benignform, inherited as a dominant, presents in adults, <strong>and</strong> amalignant one, inherited as a recessive, in infants <strong>and</strong>young children. The patients with <strong>the</strong> benign form <strong>of</strong>tensurvive to old age <strong>and</strong> present prominent otologicalsymptoms. The intermediate, endochondral portion<strong>of</strong> <strong>the</strong> otic capsule is swollen <strong>and</strong> appears as an exaggeratedlythickened form <strong>of</strong> <strong>the</strong> normal state. Globuliossei composed <strong>of</strong> groups <strong>of</strong> calcified cartilage cells arenormally present in this region, <strong>and</strong> in osteopetrosis<strong>the</strong>y are greatly increased in number <strong>and</strong> are arrangedinto a markedly thickened zone. The periosteal bone isnormal. The ossicles are <strong>of</strong> foetal shape <strong>and</strong> filled withunabsorbed, calcified cartilage. The canals for <strong>the</strong> seventh<strong>and</strong> eighth cranial nerves are greatly narrowed by<strong>the</strong> exp<strong>and</strong>ed cartilaginous <strong>and</strong> bony tissue <strong>and</strong> <strong>the</strong>sechanges are probably responsible for <strong>the</strong> characteristicsymptoms <strong>of</strong> facial palsy <strong>and</strong> hearing loss respectively[36, 74, 77].8.4.2 Membranous Labyrinth<strong>and</strong> Cranial Nerves8.4.2.1 Viral, Bacterial<strong>and</strong> Mycotic InfectionsCytomegaloviruses are DNA-containing members <strong>of</strong><strong>the</strong> herpesvirus group. General infection is frequent,an intrauterine source <strong>of</strong>ten being incriminated. Thedeveloping human ear has been thought to be particularlysusceptible to CMV infection [106] <strong>and</strong> <strong>the</strong>virus has been incriminated on clinical <strong>and</strong> virologicalgrounds as <strong>the</strong> most common cause <strong>of</strong> congenitalhearing loss [24, 25, 37, 76, 112]. In infant inner ears<strong>the</strong> endolabyrinth is mainly involved. CMV infectionis commonly seen in patients with AIDS. Thirty-ninepercent <strong>of</strong> patients with AIDS were found to have ahearing loss <strong>of</strong> sensorineural type [104]. In a study <strong>of</strong><strong>the</strong> temporal bones at autopsy <strong>of</strong> 25 patients CMV infectionwas identified in <strong>the</strong> inner ears <strong>of</strong> 5 patients by<strong>the</strong> presence <strong>of</strong> <strong>the</strong> characteristic inclusions. The inclusionswere found in <strong>the</strong> vestibular nerve in <strong>the</strong> internalcanal (Fig. 8.19), in <strong>the</strong> stria <strong>and</strong> in <strong>the</strong> saccule, utricle<strong>and</strong> lateral semicircular duct [69].It is likely, <strong>the</strong>refore, that <strong>the</strong> hearing loss in patientswith AIDS is due to cochlear CMV infection.


Ear <strong>and</strong> Temporal Bone Chapter 8 255Maternal rubella is an important factor in <strong>the</strong> genesis<strong>of</strong> congenital sensorineural hearing loss. The virus is anRNA one. In two cases <strong>the</strong> temporal bones showed inflammatorycollections at <strong>the</strong> upper end near <strong>the</strong> junctionwith Reissner’s membrane <strong>and</strong> adherent to it [29].The organ <strong>of</strong> Corti was mainly normal.In herpes zoster auris (Ramsay Hunt syndrome), <strong>the</strong>virus (<strong>the</strong> DNA herpes varicella virus) enters <strong>the</strong> inner earalong <strong>the</strong> seventh <strong>and</strong> eighth cranial nerves, presumablyfrom nerve ganglia where it lies dormant until <strong>the</strong> immunologicalstatus <strong>of</strong> <strong>the</strong> patient deteriorates. In histopathologicalstudies previously described <strong>the</strong>re were extensiveinflammatory changes mainly in those two cranialnerves serving in <strong>the</strong> transmission <strong>of</strong> <strong>the</strong> virus. Varicellazoster has also been detected in <strong>the</strong> cytoplasm <strong>and</strong> nuclei<strong>of</strong> inflammatory cells <strong>of</strong> <strong>the</strong> middle ear in two cases <strong>of</strong><strong>the</strong> Ramsay-Hunt syndrome by an immun<strong>of</strong>luorescencemethod [30]. Herpes varicella-zoster viral (VZV) DNAhas been identified, using <strong>the</strong> polymerase chain reaction,in archival celloidin-embedded temporal bone sectionsfrom two patients who clinically had Ramsay-Hunt syndrome(herpes zoster oticus) [119].A condition possibly due to viral infection in <strong>the</strong> innerear is that <strong>of</strong> Bell’s palsy, which is manifested clinicallyas a peripheral facial paralysis. The suggestion hasbeen made, with some virological support, that this conditionis <strong>the</strong> result <strong>of</strong> infection with herpes simplex virus,type 1. There have been a very small number <strong>of</strong> reports<strong>of</strong> temporal bone studies from patients with Bell’spalsy. In two cases <strong>of</strong> Bell’s palsy I studied, serial sections<strong>of</strong> <strong>the</strong> temporal bones both showed <strong>the</strong> followinghistological findings. In <strong>the</strong> genu region <strong>the</strong>re appearedto be constriction <strong>of</strong> <strong>the</strong> facial nerve by inflammatorytissue, which formed a sheath around it <strong>and</strong> encroachedon its interior. The adjacent bone showed foci <strong>of</strong> resorptionwith abundant osteoclasts (Fig. 8.20). The geniculateganglion was infiltrated by lymphocytes. In someplaces <strong>the</strong> affected facial nerve appeared severely oedematous<strong>and</strong> nerve cells were shrunken <strong>and</strong> showed aneosinophilic cytoplasm. The descending part <strong>of</strong> <strong>the</strong> facialnerve presented swelling <strong>and</strong> vacuolation <strong>of</strong> myelinsheaths with some loss <strong>of</strong> axis cylinders. These findingsare compatible with geniculate ganglionitis. In one <strong>of</strong><strong>the</strong>se cases, herpes simplex viral type 1 was demonstratedin archival paraffin-embedded sections <strong>of</strong> <strong>the</strong> affectedgeniculate ganglion by carrying out PCR followed byelectrophoresis on agarose gel [14].Pe t ro s i t i s : bacterial infections <strong>of</strong> <strong>the</strong> inner ear may involveboth <strong>the</strong> petrous bone itself <strong>and</strong> <strong>the</strong> labyrinthinestructures within it. Bacterial infection <strong>of</strong> <strong>the</strong> petrousbone is frequently derived by extension from middle earinfection. There are four possible routes by which infectionmay extend from <strong>the</strong> middle ear into <strong>the</strong> petrousbone [68]:1. Via air cells. Mastoid air cells frequently extend in<strong>the</strong> temporal bone as far as <strong>the</strong> apical region. It isFig. 8.20. Interface between geniculate ganglion <strong>and</strong> adjacentbone in a case <strong>of</strong> Bell’s palsy, showing numerous osteoclasts withHowship’s lacunae. Reproduced from Michaels <strong>and</strong> Hellquist [68]possible, <strong>the</strong>refore, that infection to <strong>the</strong> petrous apexmay extend from <strong>the</strong> middle ear by <strong>the</strong> medium <strong>of</strong>infection <strong>of</strong> air cells;2. As direct spread <strong>of</strong> <strong>the</strong> inflammatory process by bonenecrosis (osteitis);3. By extension through <strong>the</strong> bone marrow <strong>of</strong> <strong>the</strong> petrousbone (osteomyelitis);4. Along vessels <strong>and</strong> nerves.In addition to inflammatory infiltration <strong>the</strong> pathologicalprocess <strong>of</strong> petrositis comprises three main changesin <strong>the</strong> bone tissue, all <strong>of</strong> which may be seen simultaneously:(a) bone necrosis, (b) bone erosion, (c) new boneformation. Petrositis is <strong>of</strong> great importance because involvement<strong>of</strong> <strong>the</strong> labyrinth, nerves, artery, veins, meninges<strong>and</strong> cerebral tissue embedded in <strong>and</strong> surrounding<strong>the</strong> petrous bone may each cause serious symptoms, <strong>and</strong>perhaps death.Extension to <strong>the</strong> labyrinth may lead to labyrinthitiswith destruction <strong>of</strong> <strong>the</strong> organs <strong>of</strong> hearing <strong>and</strong> balance.Important nerves may be damaged. The facial nerveis at risk early on. Involvement <strong>of</strong> <strong>the</strong> trigeminal ganglion<strong>and</strong> <strong>the</strong> sixth cranial nerve lead to “Gradenigo’ssyndrome”. Extension to <strong>the</strong> jugular foramen region by<strong>the</strong> inflammatory process may cause palsy <strong>of</strong> <strong>the</strong> ninth,tenth <strong>and</strong> eleventh cranial nerves (“jugular foramensyndrome”).The wall <strong>of</strong> <strong>the</strong> internal carotid artery may becomeinflamed <strong>and</strong> this may lead to thrombosis <strong>of</strong> <strong>the</strong> vesselwith possible cerebral complications. Similarly, <strong>the</strong>lateral sinus may become thrombosed <strong>and</strong> this <strong>and</strong>/orextension <strong>of</strong> <strong>the</strong> thrombus to <strong>the</strong> superior sagittal sinusmay be associated with <strong>the</strong> somewhat arcane syndrome<strong>of</strong> otitic hydrocephalus. Spread <strong>of</strong> <strong>the</strong> infectionto <strong>the</strong> immediately adjacent cranial structures will leadto meningitis <strong>and</strong> cerebral abscess.Labyrinthitis: <strong>the</strong> source <strong>of</strong> labyrinthitis is, in manyinstances, otitis media, as with petrositis. Infection may


256 L. Michaels8enter <strong>the</strong> labyrinth by penetrating <strong>the</strong> oval or <strong>the</strong> roundwindow. An infected air cell may rupture into <strong>the</strong> labyrinthinesystem at some point <strong>of</strong> its complex periphery.Occasionally, damage to bone by <strong>the</strong> inflammation mayproduce a fistula between <strong>the</strong> middle ear <strong>and</strong> <strong>the</strong> labyrinth,usually in <strong>the</strong> lateral semicircular canal becausethis is <strong>the</strong> nearest vulnerable point to <strong>the</strong> middle ear.The latter complication takes place in most cases when acholesteatoma is present, which has <strong>the</strong> effect <strong>of</strong> stimulating<strong>the</strong> inflammatory process.Infection may also be conveyed from meningitisthrough <strong>the</strong> cochlear aqueduct <strong>and</strong> <strong>the</strong> internal auditorymeatus into <strong>the</strong> labyrinth. Sensorineural hearingloss is an important sequela <strong>of</strong> acute bacterial meningitis[66].In suppurative labyrinthitis <strong>the</strong> perilymph spaces displaya usually massive exudate <strong>of</strong> neutrophils. If <strong>the</strong> processextends to <strong>the</strong> endolymphatic spaces <strong>the</strong>re is concomitantdestruction <strong>of</strong> membranous structures <strong>and</strong> irreparabledamage to sensory epi<strong>the</strong>lia.Healing is at first by fibrosis, but later osseous repairis frequent, leading to a condition <strong>of</strong> “labyrinthitis ossificans”.In this condition <strong>the</strong> spaces <strong>of</strong> <strong>the</strong> bony labyrinthare filled in by a newer bone, which appears instriking contrast with <strong>the</strong> normal bone surrounding <strong>the</strong>bony labyrinth.Cryptococcosis is a fungus infection that usually infects<strong>the</strong> meninges. There may be extension by <strong>the</strong> organismCryptococcus ne<strong>of</strong>ormans, from <strong>the</strong> meningesalong <strong>the</strong> internal auditory meatus <strong>and</strong> <strong>the</strong>n into <strong>the</strong> cochleavia <strong>the</strong> modiolus. Such a progression was clearlypresent in two cases <strong>of</strong> AIDS with cryptococcal meningitisthat had spread to <strong>the</strong> labyrinth [69].8.4.2.2 Lesions<strong>of</strong> <strong>the</strong> Vestibular SystemThe pathology <strong>of</strong> <strong>the</strong> vestibular labyrinth has not beenas well studied as that <strong>of</strong> <strong>the</strong> cochlea <strong>and</strong> o<strong>the</strong>r parts <strong>of</strong><strong>the</strong> labyrinth. This is <strong>the</strong> result <strong>of</strong> <strong>the</strong> paucity <strong>of</strong> operativeprocedures with biopsy carried out in this area <strong>and</strong>also <strong>of</strong> <strong>the</strong> rapidity <strong>of</strong> autolysis, which takes place afterdeath so that histological study <strong>of</strong> this area is difficult.The non-neoplastic lesions listed below are described indetail by Michaels <strong>and</strong> Hellquist [68].Ototoxicity: many drugs damage <strong>the</strong> sensory epi<strong>the</strong>lia<strong>of</strong> <strong>the</strong> inner ear. The most obvious clinical effect is when<strong>the</strong> cochlear sensory cells are involved so that hearingloss results. Part <strong>of</strong> <strong>the</strong> damage produced by aminoglycosideantibiotics such as gentamycin, however, may beto <strong>the</strong> sensory epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong> cristae <strong>and</strong> maculae,producing symptoms <strong>of</strong> imbalance.Virus infection: in rubella <strong>and</strong> cytomegalovirus infectionchanges have been observed in <strong>the</strong> utricle <strong>and</strong>saccule (see above).Fig. 8.21. Hydrops <strong>of</strong> scala media <strong>of</strong> <strong>the</strong> cochlea. Reisner’s membraneis distended to such a degree that it touches <strong>the</strong> top <strong>of</strong> <strong>the</strong>scala vestibuli. Reproduced from Michaels <strong>and</strong> Hellquist [68]Bacterial infection: bacterial infection may involve<strong>the</strong> vestibular system as part <strong>of</strong> labyrinthitis. In mostbacterial infections, spread occurs from <strong>the</strong> middle earvia <strong>the</strong> oval window. A direct fistula resulting from <strong>the</strong>bone erosion <strong>of</strong> otitis media may take place leading into<strong>the</strong> lateral semicircular canal, particularly in <strong>the</strong> presence<strong>of</strong> cholesteatoma.Bone diseases: Paget’s disease frequently involves <strong>the</strong>bony vestibule <strong>and</strong> semicircular canals to a severe degree<strong>and</strong> as a result clinical symptoms referable to thissystem are likely to occur. Otosclerosis, although frequentlypresent in relation to <strong>the</strong> bony wall <strong>of</strong> <strong>the</strong> vestibule,rarely involves <strong>the</strong> membranous structures <strong>of</strong> <strong>the</strong>vestibular system so that vestibular symptoms are rarein this condition.Hydrops <strong>of</strong> <strong>the</strong> saccule, which sometimes extends to<strong>the</strong> utricle, is <strong>the</strong> major pathological feature <strong>of</strong> Ménière’sdisease <strong>and</strong> is responsible for <strong>the</strong> characteristic symptom<strong>of</strong> that disease – vertigo. The scala media <strong>of</strong> <strong>the</strong> cochleais usually distended in Ménière’s disease (Fig. 8.21), <strong>and</strong>this is <strong>the</strong> pathological basis <strong>of</strong> <strong>the</strong> hearing loss <strong>and</strong> tinnitusthat are <strong>the</strong> o<strong>the</strong>r disturbing symptoms in attacks<strong>of</strong> <strong>the</strong> disease. Saccular hydrops may also be a manifestation<strong>of</strong> syphilitic <strong>and</strong> bacterial inflammation involving<strong>the</strong> labyrinth.Positional vertigo is a very common condition inwhich vertigo is induced in <strong>the</strong> patient by alterationin <strong>the</strong> position <strong>of</strong> <strong>the</strong> head. In 1969, Schuknecht described<strong>the</strong> temporal bone findings in two cases <strong>of</strong>positional vertigo [94]. Attached to <strong>the</strong> posterior surface<strong>of</strong> <strong>the</strong> cupula <strong>of</strong> <strong>the</strong> left posterior semicircularcanal in each <strong>of</strong> <strong>the</strong> cases was a basophilically stainedhomogeneous deposit. On <strong>the</strong> basis <strong>of</strong> <strong>the</strong>se cases,Schuknecht has built up an explanation <strong>of</strong> <strong>the</strong> symptomatology<strong>of</strong> positional vertigo postulating that <strong>the</strong>calcific material derived from otoconia in a degeneratedutricular macula will descend by gravity along


Ear <strong>and</strong> Temporal Bone Chapter 8 257Fig. 8.22. Vestibular schwannoma in a microsliced temporalbone. The neoplasm is arising from <strong>the</strong> vestibular division <strong>of</strong> <strong>the</strong>eighth nerve <strong>and</strong> compressing <strong>the</strong> cochlear division. Note <strong>the</strong>granular deposit lining <strong>the</strong> cochlea. Reproduced from Michaels<strong>and</strong> Hellquist [68]Fig. 8.23. Small vestibular schwannoma. It arises from <strong>the</strong> vestibulardivision <strong>of</strong> <strong>the</strong> eighth nerve in <strong>the</strong> region <strong>of</strong> <strong>the</strong> glial neurilemmaljunction <strong>and</strong> causes only a small indentation <strong>of</strong> <strong>the</strong> bonywall <strong>of</strong> <strong>the</strong> internal canal. There is exudate in <strong>the</strong> vestibule, but notin <strong>the</strong> cochlea. Reproduced from Michaels <strong>and</strong> Hellquist [68]<strong>the</strong> endolymph <strong>and</strong> form on <strong>the</strong> crista <strong>of</strong> <strong>the</strong> posteriorsemicircular canal, <strong>the</strong> lowest region <strong>of</strong> <strong>the</strong> labyrinthinesensory epi<strong>the</strong>lium. This ingenious <strong>the</strong>ory hasattracted much interest <strong>and</strong> <strong>the</strong> term “cupulolithiasis”is nowadays frequently used as a synonym for positionalvertigo.8.4.2.3 Tumours<strong>and</strong> Tumour-Like LesionsThe most important neoplasm <strong>of</strong> <strong>the</strong> vestibular systemis schwannoma (acoustic neuroma, ICD-O:9560/0) <strong>of</strong> <strong>the</strong>vestibular division <strong>of</strong> <strong>the</strong> eighth cranial nerve. This doesnot usually invade <strong>the</strong> vestibule, but may do so in cases<strong>of</strong> neur<strong>of</strong>ibromatosis [2] (see below). The saccule <strong>and</strong>utricle show an exudate <strong>of</strong> proteinaceous fluid in <strong>the</strong>presence <strong>of</strong> a schwannoma <strong>of</strong> <strong>the</strong> internal auditory meatus.Ano<strong>the</strong>r much rarer neoplasm is <strong>the</strong> low-grade adenocarcinoma<strong>of</strong> <strong>the</strong> endolymphatic sac (endolymphaticsac tumour; see below). Metastatic deposits are unusual;invasion <strong>of</strong> <strong>the</strong> vestibular system from <strong>the</strong> internal auditorymeatus by way <strong>of</strong> <strong>the</strong> vestibular nerve may occur inmetastatic neoplasm or in carcinoma <strong>of</strong> <strong>the</strong> middle ear(see above).Vestibular schwannoma is stated to arise most commonlyat <strong>the</strong> glial-neurilemmal junction <strong>of</strong> <strong>the</strong> eighthnerve, which is usually within <strong>the</strong> internal auditory meatus.In one study <strong>of</strong> five temporal bones with small vestibularschwannomas, <strong>the</strong> tumour arose more peripherally,however [128]. When seen at surgery or autopsyvestibular schwannoma in most cases is found to occupya much greater part <strong>of</strong> <strong>the</strong> nerve. Usually it is <strong>the</strong> vestibulardivision <strong>of</strong> <strong>the</strong> nerve that is affected; in a few <strong>the</strong> cochleardivision is <strong>the</strong> source <strong>of</strong> <strong>the</strong> neoplasm (Fig. 8.22).Growth takes place from origin, both centrally onto <strong>the</strong>cerebellopontine angle, <strong>and</strong> distally along <strong>the</strong> canal.Vestibular schwannoma is usually unilateral, but may bebilateral (see below).The neoplasm may grow slowly for years withoutcausing symptoms <strong>and</strong> may be first diagnosed only atpost-mortem where it has been found in about 1 in 220consecutive adults [57]. Although it arises on <strong>the</strong> vestibularbranch <strong>of</strong> <strong>the</strong> eighth cranial nerve, hearing loss <strong>and</strong>tinnitus are early symptoms produced by involvement <strong>of</strong><strong>the</strong> cochlear division <strong>of</strong> <strong>the</strong> nerve; in <strong>the</strong> later stages vertigo<strong>and</strong> abnormal caloric <strong>and</strong> electronystagmographicresponses develop from damage to <strong>the</strong> vestibular divisionitself. Surgical removal may be carried out by drillingfrom <strong>the</strong> external canal through <strong>the</strong> temporal bone,by craniotomy <strong>and</strong> <strong>the</strong> middle fossa approach to <strong>the</strong> internalauditory meatus, or by stereotactically guidedgamma knife surgery.The neoplasm, seen grossly, is <strong>of</strong> variable size <strong>and</strong> <strong>of</strong>round or oval shape. Small tumours ei<strong>the</strong>r do not widen<strong>the</strong> canal at all or produce only a small indentationin <strong>the</strong> bone (Fig. 8.23). The larger tumours <strong>of</strong>ten have amushroom shape with two components, <strong>the</strong> stalk – anelongated part in <strong>the</strong> canal – <strong>and</strong> an exp<strong>and</strong>ed part in<strong>the</strong> region <strong>of</strong> <strong>the</strong> cerebellopontine angle. The bone <strong>of</strong> <strong>the</strong>internal auditory canal is widened funnel-wise as <strong>the</strong>neoplasm grows. The tumour surface is smooth <strong>and</strong> lobulated.The cut surface is yellowish, <strong>of</strong>ten with areas <strong>of</strong>haemorrhage <strong>and</strong> cysts. The vestibular division <strong>of</strong> <strong>the</strong>eighth nerve may be identified on <strong>the</strong> surface <strong>of</strong> <strong>the</strong> tumour.Acoustic neuroma has <strong>the</strong> features <strong>of</strong> a neoplasm <strong>of</strong>Schwann cells showing Antoni A <strong>and</strong> Antoni B areas.Antoni A areas display spindle cells closely packed toge<strong>the</strong>rwith palisading <strong>of</strong> nuclei. Verocay bodies, whichmay be present in <strong>the</strong> Antoni A areas, are whorled formations<strong>of</strong> palisaded tumour cells. The degree <strong>of</strong> cellularity<strong>of</strong> <strong>the</strong> neoplasm can be high or low. The spindlecells frequently are moderately pleomorphic, but mitotic


258 L. Michaels8Fig. 8.24. Lipoma <strong>of</strong> <strong>the</strong> internal auditory canal. Note nervebranch passing through adipose tissue near <strong>the</strong> bottom. Reproducedfrom Michaels <strong>and</strong> Hellquist [68]figures are unusual. The presence <strong>of</strong> pleomorphism doesnot denote a malignant tendency. Antoni B areas, probablya degenerated form <strong>of</strong> <strong>the</strong> Antoni A pattern, show aloose reticular pattern, sometimes with histiocytic proliferation.Thrombosis <strong>and</strong> necrosis may be present insome parts <strong>of</strong> <strong>the</strong> neoplasm. A mild degree <strong>of</strong> invasion<strong>of</strong> modiolus or vestibule along cochlear or vestibularnerve branches may be present even in solitary vestibularschwannomas. Granular or homogeneous fluid exudateis usually present in <strong>the</strong> perilymphatic spaces <strong>of</strong><strong>the</strong> cochlea <strong>and</strong> vestibule. This may arise as a result <strong>of</strong>pressure by <strong>the</strong> neoplasm on veins draining <strong>the</strong> cochlea<strong>and</strong> vestibule in <strong>the</strong> internal auditory meatus. Hydrops<strong>of</strong> <strong>the</strong> endolymphatic system may occur (see above) <strong>and</strong>in larger tumours <strong>the</strong>re is atrophy <strong>of</strong> spiral ganglion cells<strong>and</strong> nerve fibres in <strong>the</strong> basilar membrane.The tumour is benign <strong>and</strong> usually grows slowly. Serioussymptoms <strong>and</strong> even death may occur, however, dueto damage to cerebral structures if <strong>the</strong> neoplasm growsto a large size.Neur<strong>of</strong>ibromatosis 2 (Bilateral Vestibular Schwannoma,ICD-O:9540/1): bilateral vestibular schwannomaacoustic neuroma (neur<strong>of</strong>ibromatosis 2, NF2) is, unlikeneur<strong>of</strong>ibromatosis 1 (von Recklinghausen’s disease), notassociated with large numbers <strong>of</strong> cutaneous neur<strong>of</strong>ibromas<strong>and</strong> cafe-au-lait spots, but <strong>the</strong> temporal bone locality<strong>of</strong> <strong>the</strong> neural tumour <strong>and</strong> its bilaterality are inherited asan autosomal dominant trait. This condition has been relatedto a gene localised near <strong>the</strong> centre <strong>of</strong> <strong>the</strong> long arm <strong>of</strong>chromosome 22. At autopsy <strong>of</strong> cases <strong>of</strong> neur<strong>of</strong>ibromatosis2, neural neoplasms are present in both eighth nerves<strong>and</strong> o<strong>the</strong>r central nerves. There are <strong>of</strong>ten many smallschwannomas <strong>and</strong> collections <strong>of</strong> cells <strong>of</strong> neur<strong>of</strong>ibromatous<strong>and</strong> meningiomatous appearance growing on cranialnerves <strong>and</strong> on <strong>the</strong> meninges in <strong>the</strong> vicinity <strong>of</strong> <strong>the</strong> vestibularschwannomas <strong>and</strong> sometimes even intermixedwith <strong>the</strong>m. The NF2 tumours are histologically similarto those <strong>of</strong> <strong>the</strong> single tumours except that <strong>the</strong> former havemore Verocay bodies <strong>and</strong> more foci <strong>of</strong> high cellularity.The NF2 tumours are more invasive, however, tending toinfiltrate <strong>the</strong> cochlea <strong>and</strong> vestibule more deeply.As with all schwannomas, <strong>the</strong> strongest <strong>and</strong> mostconsistent immunohistochemical reaction is <strong>the</strong> positivitydisplayed when staining with a polyclonal antibodyagainst S-100 protein is carried out. The vimentinmarker is also usually positive. These findings arecommon to both unilateral vestibular schwannoma <strong>and</strong><strong>the</strong> schwannomas <strong>of</strong> NF2. Glial fibrillary acidic protein<strong>and</strong> neuron-specific enolase markers are also sometimespositive; <strong>the</strong> tumours are consistently negative forCD34, a marker widely used for <strong>the</strong> diagnosis <strong>of</strong> solitaryfibrous tumours, unless <strong>the</strong> vestibular schwannoma iswidely degenerated [117].An antibody against Ki67 (MIB1 in paraffin sections)has been utilised in a number <strong>of</strong> investigations to determinewhe<strong>the</strong>r <strong>the</strong> degree <strong>of</strong> positivity with this proliferationmarker can be related to <strong>the</strong> clinical activity <strong>of</strong> <strong>the</strong>tumour. It has been demonstrated that tumours 18 mmor smaller in diameter have lower proliferation indices<strong>and</strong> growth rates, compared with tumours larger than18 mm [6]. The degree <strong>of</strong> labelling with <strong>the</strong> proliferationmarker is higher in cases <strong>of</strong> NF2 than in those <strong>of</strong> solitaryvestibular schwannoma [2].Men i ng ioma s (ICD-O:9530/0) are usually intracranialmasses. They arise from arachnoid villi, which aresmall protrusions <strong>of</strong> <strong>the</strong> arachnoid membranes into <strong>the</strong>venous sinuses. Arachnoid villi may be found in parts <strong>of</strong><strong>the</strong> temporal bone, including <strong>the</strong> inner ear, <strong>and</strong> on occasionmeningiomas may arise from <strong>the</strong>se structures as primaryneoplasms <strong>of</strong> <strong>the</strong> inner ear region. The most likelyposition for a primary inner ear meningioma is in <strong>the</strong>wall <strong>of</strong> <strong>the</strong> internal auditory meatus, where arachnoid villiare normally frequent. The histological appearances <strong>of</strong>a meningioma are those <strong>of</strong> a tumour with a whorled arrangement<strong>of</strong> cells: meningo<strong>the</strong>liomatous if <strong>the</strong> tumourcells appear epi<strong>the</strong>lioid, psammomatous if calcification <strong>of</strong><strong>the</strong> whorled masses is prominent <strong>and</strong> fibroblastic if <strong>the</strong>tumour cells resemble fibroblasts. Meningiomas as wellas acoustic neuromas may appear in <strong>the</strong> inner ear in <strong>the</strong>NF2 syndrome. The meningioma is a slowly growing tumour<strong>of</strong> <strong>the</strong> temporal bone that has had a reputation forcomplete benignity. In <strong>the</strong> temporal bone, however, middleear meningioma sometimes has a strong propensitytowards local recurrence <strong>and</strong> invasion (see above).Li p o m a s (ICD-O:8850/0) <strong>of</strong> <strong>the</strong> internal auditory canal<strong>and</strong> cerebellopontine angle are rare tumours thatmay be confused clinically with <strong>the</strong> much commonervestibular schwannoma. On magnetic resonance usingfat-suppressed T1-weighted images after gadoliniumenhancement, however, this tumour displays characteristics<strong>of</strong> adipose tissue ra<strong>the</strong>r than those <strong>of</strong> schwannoma.There may be erosion <strong>of</strong> <strong>the</strong> walls <strong>of</strong> <strong>the</strong> internal auditorycanal as with vestibular schwannoma, <strong>and</strong> lipomamay appear similar to <strong>the</strong> latter at operation. Since


Ear <strong>and</strong> Temporal Bone Chapter 8 259Fig. 8.25. Low-grade adenocarcinoma <strong>of</strong> probable endolymphaticsac origin showing a papillary pattern. The epidermoid epi<strong>the</strong>liumon <strong>the</strong> right is probably that <strong>of</strong> <strong>the</strong> external auditory canal towhich <strong>the</strong> tumour had extended. Reproduced from Michaels <strong>and</strong>Hellquist [68]Fig. 8.26. Low-grade adenocarcinoma <strong>of</strong> probable endolymphaticsac origin showing a thyroid-like gl<strong>and</strong>ular pattern from ano<strong>the</strong>rarea <strong>of</strong> <strong>the</strong> tumour shown in Fig. 8.25. Reproduced from Michaels<strong>and</strong> Hellquist [68]<strong>the</strong> seventh <strong>and</strong> eighth cranial nerves or <strong>the</strong>ir branches(Fig. 8.24) may pass through <strong>the</strong> lesion <strong>and</strong> <strong>the</strong>ir integritybe damaged by removal <strong>of</strong> <strong>the</strong> tumour it is recommendedthat diagnosis be made whenever <strong>the</strong> possibility<strong>of</strong> this neoplasm is suspected at operation by examination<strong>of</strong> frozen sections. If a diagnosis <strong>of</strong> lipomais made in this way <strong>the</strong> tumour should not be resected,since its fur<strong>the</strong>r growth does not constitute a threat tovital structures [102].Low-grade adenocarcinoma <strong>of</strong> probable endolymphaticsac origin ( endolymphatic sac tumour): <strong>the</strong>re is evidence<strong>of</strong> <strong>the</strong> existence <strong>of</strong> an epi<strong>the</strong>lial neoplasm <strong>of</strong> <strong>the</strong> endolymphaticsystem, mainly in <strong>the</strong> endolymphatic sac [35,38, 41]. Although <strong>of</strong> bl<strong>and</strong> histological appearance <strong>and</strong><strong>of</strong> slow growth <strong>the</strong> neoplasm seems to have considerableinvasive capacity <strong>and</strong> <strong>the</strong>refore <strong>the</strong> term “low-grade adenocarcinoma<strong>of</strong> probable endolymphatic sac origin” hasbeen applied. O<strong>the</strong>r terms, such as endolymphatic sac tumour<strong>and</strong> Heffner’s tumour, are also in use. Some caseshave presented bilateral neoplasms <strong>of</strong> <strong>the</strong> same type<strong>and</strong> some have also been associated with von Hippel-Lindaudisease [64]. The course <strong>of</strong> <strong>the</strong> tumour’s growth mayextend over many years. Tinnitus or vertigo, similar oridentical to <strong>the</strong> symptoms <strong>of</strong> Ménière’s disease, are presentin about one-third <strong>of</strong> patients. It is presumed that earlyobstruction <strong>of</strong> <strong>the</strong> endolymphatic sac leads to hydrops<strong>of</strong> <strong>the</strong> endolymphatic system <strong>of</strong> <strong>the</strong> labyrinth <strong>and</strong> so to<strong>the</strong> Ménière’s symptoms. Imaging reveals a lytic temporalbone lesion, appearing to originate from <strong>the</strong> regionbetween <strong>the</strong> internal auditory canal <strong>and</strong> sigmoid sinus(which is <strong>the</strong> approximate position <strong>of</strong> <strong>the</strong> endolymphaticsac). There is usually prominent extension into <strong>the</strong> posteriorcranial cavity <strong>and</strong> invasion <strong>of</strong> <strong>the</strong> middle ear.In most cases <strong>the</strong> tumour has a papillary-gl<strong>and</strong>ularappearance, <strong>the</strong> papillary proliferation being linedby a single row <strong>of</strong> low cuboidal cells. The vascular nature<strong>of</strong> <strong>the</strong> papillae in some cases has given <strong>the</strong> tumoura histological resemblance to choroid plexus papilloma(Fig. 8.25). In some cases <strong>the</strong> tumour also shows areas<strong>of</strong> dilated gl<strong>and</strong>s containing secretion that has some resemblanceto colloid <strong>and</strong> under <strong>the</strong>se circumstances <strong>the</strong>lesion may resemble papillary adenocarcinoma <strong>of</strong> <strong>the</strong>thyroid (Fig. 8.26). Such thyroid-like areas may evendominate <strong>the</strong> histological pattern. A few cases show aclear cell predominance resembling carcinoma <strong>of</strong> <strong>the</strong>kidney. On immunohistochemistry <strong>the</strong> epi<strong>the</strong>lial cells<strong>of</strong> this neoplasm contain antigens <strong>of</strong> cytokeratins. Sometumours contain glial fibrillary acidic protein. Thyroglobulinis always absent.It seems possible that many cases <strong>of</strong> <strong>the</strong> so-called“aggressive papillary middle ear tumours” may be lowgradeadenocarcinomas <strong>of</strong> <strong>the</strong> endolymphatic sac wi<strong>the</strong>xtension <strong>of</strong> <strong>the</strong> neoplasm to <strong>the</strong> middle ear (see alsoSect. 8.3.2.3) [31]. Not all <strong>of</strong> such tumours may arise in<strong>the</strong> endolymphatic sac [89].The histological appearances <strong>of</strong> low-grade adenocarcinomas<strong>of</strong> probable endolymphatic sac origin are indeedin keeping with <strong>the</strong> normal histological structure<strong>of</strong> <strong>the</strong> endolymphatic sac, which is lined by a papillarycolumnar epi<strong>the</strong>lial layer.Cholesteatoma (epidermoid cyst) usually presentswith symptoms relating to its involvement <strong>of</strong> <strong>the</strong> seventh<strong>and</strong> eighth cranial nerves in <strong>the</strong> cerebellopontineangle. The histological appearance is similar to that <strong>of</strong>middle ear cholesteatoma (see above). It is probably <strong>of</strong>congenital origin, but no cell rest has been discoveredfrom which it might arise.Cholesterol granuloma is a lesion <strong>of</strong> <strong>the</strong> petrous apexwith <strong>the</strong> typical features <strong>of</strong> cholesterol granuloma asseen in <strong>the</strong> middle ear <strong>and</strong> mastoid in chronic otitis media,<strong>and</strong> has been identified in recent years with increasingfrequency. At operation it appears cystic, <strong>the</strong> contentsbeing altered blood <strong>and</strong> cholesterol clefts with aforeign body giant cell reaction.


262 L. Michaels879. Nager GT (1975) Paget’s disease <strong>of</strong> <strong>the</strong> temporal bone. AnnOtol Rhinol Laryngol 84 [Suppl] 22:3–3280. Nager GT, Kennedy DW, Kopstein E (1982) Fibrous dysplasia:a review <strong>of</strong> <strong>the</strong> disease <strong>and</strong> its manifestations in <strong>the</strong> temporalbone. Ann Otol Rhinol Laryngol 91:Suppl 9281. Nakhleh RE, Swanson PE, Dehner LP (1991) Juvenile (embryonal<strong>and</strong> alveolar) rhabdomyosarcoma <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck in adults. A clinical, pathologic, <strong>and</strong> immunohistochemicalstudy <strong>of</strong> 12 cases. Cancer 67:1019–102482. Nelson EG, Kratz RC (1993) Sebaceous choristoma <strong>of</strong> <strong>the</strong>middle ear. Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 108:372-37383. Nyrop M, Grantved A (2002) Cancer <strong>of</strong> <strong>the</strong> external auditorycanal. Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 128:834–83784. Ophir D (1991) Familial multicentric paragangliomas in achild. J Laryngol Otol 105:376–38085. Ostfeld E, Segal M, Czernobilsky B (1982) External otitis:early histopathologic changes <strong>and</strong> pathogenic mechanism.Laryngoscope 91:965–97086. Palmer JM, Coker NJ, Harper RL (1989) Papillary adenoma<strong>of</strong> <strong>the</strong> temporal bone in von Hippel-Lindau disease. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 100:64–6887. Persaud R, Haji<strong>of</strong>f D, Thevasagayam MS, Wareing MJ, WrightA (2004) Keratosis obturans <strong>and</strong> external ear canal cholesteatoma:how <strong>and</strong> why we should distinguish between <strong>the</strong>seconditions. Clin Otolaryngol 29:1–588. Polinsky MN, Brunberg JA, McKeever PE, S<strong>and</strong>ler HM, TelianS, Ross D (1994) Aggressive papillary middle ear tumors:a report <strong>of</strong> two cases with review <strong>of</strong> <strong>the</strong> literature. Neurosurgery35:493–497; discussion 49789. Pollak A, Bohmer A, Spycher M, Fisch U (1995) Are papillaryadenomas endolymphatic sac tumors? Ann Otol Rhinol Laryngol104:613–61990. Pou AM, Vrabec JT (2002) Inverting papilloma <strong>of</strong> <strong>the</strong> temporalbone. Laryngoscope 112:140–14291. Ramirez-Camacho R, Vicente J, Garcia Berrocal JR, Ramony Cajal S (1999) Fibro-osseous lesions <strong>of</strong> <strong>the</strong> external auditorycanal. Laryngoscope 109:488–49192. Ribe A, Fern<strong>and</strong>ez PL, Ostertarg H, Claros P, Bombi JA, PalacinA, Cardesa A (1997) Middle-ear adenoma (MEA): a report<strong>of</strong> two cases, one with predominant “plasmacytoid” features.Histopathology 30:359–36493. Roberts WH, Dinges DL, Hanly MG (1993) Inverted papilloma<strong>of</strong> <strong>the</strong> middle ear. Ann Otol Rhinol Laryngol 102:890–89294. Schuknecht H (1969) Cupolithiasis. Arch Otolaryngol90:765–77895. Schuknecht HF (1993) <strong>Pathology</strong> <strong>of</strong> <strong>the</strong> ear, 2nd edn. Lea &Febiger, Philadelphia, pp 209–21096. Schuller DE, Conley JJ, Goodman JH, Clausen KP, Miller WJ(1983) Primary adenocarcinoma <strong>of</strong> <strong>the</strong> middle ear. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 91:280–28397. Schwaber MK, Glasscock ME, Jackson CG, Smith PG (1984)Diagnosis <strong>and</strong> management <strong>of</strong> catecholamine secreting glomustumors. Laryngoscope 94:1008–101598. Seshul MJ, Eby TL, Crowe DR, Peters GE (1995) Nasal invertedpapilloma with involvement <strong>of</strong> middle ear <strong>and</strong> mastoid.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 121:1045–104899. Shah JP, Kraus DH, Dubner S, Sarkar S (1991) Patterns <strong>of</strong> regionallymph node metastases from cutaneous melanomas <strong>of</strong><strong>the</strong> head <strong>and</strong> neck. Am J Surg 162:320–323100. Siedentop KH, Jeantet C (1961) Primary adenocarcinoma <strong>of</strong><strong>the</strong> middle ear. Report <strong>of</strong> three cases. Ann Otol Rhinol Laryngol.70:719–733101. Shpitzer T, Stern Y, Cohen O, Levy R, Segal K, Feinmesser R(1993) Malignant external otitis in nondiabetic patients. AnnOtol Rhinol Laryngol 102:870–872102. Singh SP, Cottingham SL, Slone W, Boesel CP, Welling DB,Yates AJ (1996) Lipomas <strong>of</strong> <strong>the</strong> internal auditory canal. ArchPathol Lab Med 120:681–683103. Soucek S, Michaels L (1993) Keratosis <strong>of</strong> <strong>the</strong> tympanic membrane<strong>and</strong> deep external canal: a defect <strong>of</strong> auditory epi<strong>the</strong>lialmigration. Eur Arch Otorhinolaryngol 250:140–142104. Soucek S, Michaels L (1996) The ear in <strong>the</strong> acquired immunodeficiencysyndrome. II. Clinical <strong>and</strong> audiologic investigation.Am J Otol 17:35–39105. Stafford ND, Frootko NJ (1986) Verrucous carcinoma in <strong>the</strong>external auditory canal. Am J Otol 7:443–445106. Stagno S, Reynolds DW, Amos CS, Dahle AJ, McCollister FP,Mohindra I, Ermocilla R, Alford CA (1977) Auditory <strong>and</strong> visualdefects resulting from symptomatic <strong>and</strong> subclinical congenitalcytomegaloviral <strong>and</strong> toxoplasma infections. Pediatrics59:669–678107. Staibano S, Boscaino A, Salvatore G, Orabona P, PalombiniL, De Rosa G A (1996) The prognostic significance <strong>of</strong> tumorangiogenesis in nonaggressive <strong>and</strong> aggressive basal cell carcinoma<strong>of</strong> <strong>the</strong> human skin. Hum Pathol 27:695–700108. Stanley MW, Horwitz CA, Levinson RM, Sibley RK (1987)Carcinoid tumors <strong>of</strong> <strong>the</strong> middle ear. Am J Clin Pathol 87:592–600109. Stendel R, Suess O, Prosenc N, Funk T, Brock M (1998) Neoplasm<strong>of</strong> endolymphatic sac origin: clinical, radiological <strong>and</strong>pathological features. Acta Neurochir (Wien) 140:1083–1087110. Stone DM, Berktold RE, Ranganathan C, Wiet RJ (1987) Invertedpapilloma <strong>of</strong> <strong>the</strong> middle ear <strong>and</strong> mastoid. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 97:416–418111. Stone HE, Lipa M, Bell RD (1975) Primary adenocarcinoma<strong>of</strong> <strong>the</strong> middle ear. Arch Otolaryngol 101:702–705112. Strauss M (1990) Human cytomegalovirus labyrinthitis. AmJ Otolaryngol 11:292–298113. Sudh<strong>of</strong>f H, Bujia J, Fisselereckh<strong>of</strong>f A, Holly A, Schuflake C,Hildmann H (1995) Expression <strong>of</strong> a cell-cycle-associated nuclearantigen (MIB1) in cholesteatoma <strong>and</strong> auditory meatalskin. Laryngoscope 105:1227–1231114. Sudh<strong>of</strong>f H, Bujia J, Holly A, Kim C, Fischler-Eckh<strong>of</strong>f (1994)Functional characterization <strong>of</strong> middle ear mucosa residuesin cholesteatomatous samples. Am J Otol 15:217–221115. Sudh<strong>of</strong>f H, Fisseler-Eckh<strong>of</strong>f A, Stark T, Borkowski G, LuckhauptH, Cooper J, Michaels L (1997) Argyrophilic nucleolarorganizer regions in auditory meatal skin <strong>and</strong> middle earcholesteatoma. Clin Otolaryngol 22:545–548116. Thompson LD, Bouffard JP, S<strong>and</strong>berg GD, Mena H (2003)Primary ear <strong>and</strong> temporal bone meningiomas: a clinicopathologicstudy <strong>of</strong> 36 cases with a review <strong>of</strong> <strong>the</strong> literature. ModPathol 16:236–245117. Tosaka M, Hirato J, Miyagishima T, Saito N, Nakazato Y, SasakiT (2002) Calcified vestibular schwannoma with unusualhistological characteristics – positive immunoreactivity forCD-34 antigen. Acta Neurochir (Wien) 144:395–399118. Tysome JR, Harcourt J, Patel MC, S<strong>and</strong>ison A, Michaels L(2006) Aggressive papillary tumour <strong>of</strong> <strong>the</strong> middle ear. Ear,Nose <strong>and</strong> Throat Journal, in press119. Wackym PA (1997) Molecular temporal bone pathology. II.Ramsay Hunt syndrome (herpes zoster oticus). Laryngoscope107:1165–1175120. Wassef M, Kanavaros P, Polivka M, Nemeth J, Monteil JP,Frachet B, Tran Ba Huy P (1989) Middle ear adenoma. A tumordisplaying mucinous <strong>and</strong> neuroendocrine differentiation.Am J Surg Pathol 13:838–847121. Weinroth SE, Schessel D, Tuazon CU (1992) Malignant otitisexterna in AIDS patients: case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.Ear Nose Throat J 73:772–774122. Wells MD, Michaels L (1983) Role <strong>of</strong> retraction pockets incholesteatoma formation. Clin Otolaryngol 8:39–45123. Wells M, Michaels L (1984) “Malignant otitis externa”: amanifestation <strong>of</strong> chronic otitis media with complications?Clin Otolaryngol 9:131 (abstract)124. Wenig BM (1996) Schneiderian-type mucosal papillomas<strong>of</strong> <strong>the</strong> middle ear <strong>and</strong> mastoid. Ann Otol Rhinol Laryngol105:226–233125. Wetli CV, Pardo V, Millard M, Gersdon K (1972) Tumors <strong>of</strong>ceruminous gl<strong>and</strong>s. Cancer 29:1169–1178126. Wiatrak BJ, Pensak ML (1989) Rhabdomyosarcoma <strong>of</strong> <strong>the</strong>ear <strong>and</strong> temporal bone. Laryngoscope 99:1188–1192127. Wright CG, Meyerh<strong>of</strong>f WL, Burns DK (1985) Middle ear cholesteatoma:an animal model. Am J Otolaryngol 6:327–341128. Xenellis JE, Linthicum FH Jr (2003) On <strong>the</strong> myth <strong>of</strong> <strong>the</strong> glial/schwann junction (Obersteiner-Redlich zone): origin <strong>of</strong> vestibularnerve schwannomas. Otol Neurotol 24:1


Chapter 9Cysts <strong>and</strong> Unknown Primary<strong>and</strong> Secondary Tumours <strong>of</strong> <strong>the</strong> <strong>Neck</strong>,<strong>and</strong> <strong>Neck</strong> DissectionM. A Luna · K. Pineda-Daboin9Contents9.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 2649.2. Anatomy . . . . . . . . . . . . . . . . . . . . . . . . . . 2649.2.1 Triangles <strong>of</strong> <strong>the</strong> <strong>Neck</strong> . . . . . . . . . . . . . . . . . . . 2649.2.2 Lymph Node Regions <strong>of</strong> <strong>the</strong> <strong>Neck</strong> . . . . . . . . . . . . 2649.3 Cysts <strong>of</strong> <strong>the</strong> <strong>Neck</strong> . . . . . . . . . . . . . . . . . . . . . 2649.3.1 Developmental Cysts . . . . . . . . . . . . . . . . . . . 2659.3.1.1 Branchial Cleft Cysts, Sinuses <strong>and</strong> Fistulae . . . . . . 2659.3.2 Branchiogenic Carcinoma . . . . . . . . . . . . . . . . 2679.3.3 Thyroglossal Duct Cyst <strong>and</strong> Ectopic Thyroid . . . . . 2689.3.4 Cervical Thymic Cyst . . . . . . . . . . . . . . . . . . . 2699.3.5 Cervical Parathyroid Cyst . . . . . . . . . . . . . . . . 2709.3.6 Cervical Bronchogenic Cyst . . . . . . . . . . . . . . . 2709.3.7 Dermoid Cyst . . . . . . . . . . . . . . . . . . . . . . . 2719.3.8 Unclassified Cervical Cyst . . . . . . . . . . . . . . . . 2719.3.9 Non-Developmental Cysts . . . . . . . . . . . . . . . . 2719.3.9.1 Ranula . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2719.3.9.2 Laryngocele . . . . . . . . . . . . . . . . . . . . . . . . 2719.4 Cystic Neoplasms . . . . . . . . . . . . . . . . . . . . . 2729.4.1 Cystic Hygroma <strong>and</strong> Lymphangioma . . . . . . . . . . 2729.4.2 Haemangioma . . . . . . . . . . . . . . . . . . . . . . . 2729.4.3 Teratoma . . . . . . . . . . . . . . . . . . . . . . . . . . 2729.4.4 Cervical Salivary Gl<strong>and</strong> Cystic Neoplasms . . . . . . 2739.4.5 Miscellaneous Lesions . . . . . . . . . . . . . . . . . . 2739.5 Paraganglioma . . . . . . . . . . . . . . . . . . . . . . . 2739.6 Unknown Primary<strong>and</strong> Secondary Tumours . . . . . . . . . . . . . . . . . 2749.6.1 Definition . . . . . . . . . . . . . . . . . . . . . . . . . . 2749.6.2 Clinical Features . . . . . . . . . . . . . . . . . . . . . . 2759.6.3 Search for <strong>the</strong> Primary Tumour . . . . . . . . . . . . . 2759.6.4 Common Location <strong>of</strong> <strong>the</strong> Primary Tumour . . . . . . 2769.6.5 Histologic Type <strong>of</strong> Metastases<strong>and</strong> Immunohistochemical Features . . . . . . . . . . 2769.6.6 Differential Diagnosis . . . . . . . . . . . . . . . . . . . 2769.6.7 Treatment <strong>and</strong> Results . . . . . . . . . . . . . . . . . . 2789.7 <strong>Neck</strong> Dissection . . . . . . . . . . . . . . . . . . . . . . 2789.7.1 Classification <strong>of</strong> <strong>Neck</strong> Dissections . . . . . . . . . . . 2789.7.2 Gross Examination<strong>of</strong> <strong>Neck</strong> Dissection Surgical Specimens . . . . . . . . 2799.7.3 Histologic Evaluation <strong>of</strong> <strong>Neck</strong> Dissection . . . . . . . 279References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 280


264 M. A Luna · K. Pineda-Daboin9.1 IntroductionThe neck connects <strong>the</strong> organs <strong>of</strong> <strong>the</strong> head with those <strong>of</strong><strong>the</strong> thorax. It contains important anatomic structures,including blood <strong>and</strong> lymphatic vessels, nerves <strong>and</strong> paraganglia,muscles <strong>and</strong> vertebrae, <strong>and</strong> numerous lymphnodes, in addition to parenchymatous gl<strong>and</strong>s, salivary,thyroid, <strong>and</strong> parathyroid. The neck also contains organs<strong>of</strong> <strong>the</strong> upper aerodigestive tract: larynx, hypopharynx<strong>and</strong> segments <strong>of</strong> <strong>the</strong> oesophagus <strong>and</strong> trachea.The fact that a neck mass can originate in any <strong>of</strong> <strong>the</strong>cervical structures means that a host <strong>of</strong> disorders challenges<strong>the</strong> diagnostic ability <strong>of</strong> <strong>the</strong> surgical pathologist.The differential diagnosis <strong>of</strong> a neck mass includes developmental,inflammatory, benign <strong>and</strong> malignant neoplasticlesions. The purpose <strong>of</strong> this chapter is to review<strong>the</strong> pathology <strong>and</strong> diagnosis <strong>of</strong> cervical cysts. Occultprimary tumours <strong>of</strong> <strong>the</strong> neck <strong>and</strong> neck dissection alsoare discussed.99.2 Anatomy9.2.1 Triangles<strong>of</strong> <strong>the</strong> <strong>Neck</strong>It is customary to divide <strong>the</strong> neck into two large triangles,<strong>the</strong> anterior cervical triangle <strong>and</strong> <strong>the</strong> posteriorcervical triangle. The anterior triangle is bounded by<strong>the</strong> midline <strong>of</strong> <strong>the</strong> neck, <strong>the</strong> anterior border <strong>of</strong> <strong>the</strong> sternocleidomastoidmuscle, <strong>and</strong> <strong>the</strong> inferior border <strong>of</strong> <strong>the</strong>m<strong>and</strong>ible. The posterior triangle is bounded by <strong>the</strong> anteriormargin <strong>of</strong> <strong>the</strong> trapezius muscle, <strong>the</strong> posterior border<strong>of</strong> <strong>the</strong> sternocleidomastoid muscle <strong>and</strong> <strong>the</strong> clavicle.The anterior cervical triangle can be fur<strong>the</strong>r subdividedinto four lesser triangles (submental, subm<strong>and</strong>ibular,superior carotid <strong>and</strong> inferior carotid) <strong>and</strong> <strong>the</strong> posteriortriangle into two (occipital <strong>and</strong> supraclavicular)<strong>the</strong> boundaries <strong>of</strong> which are described in greater detailin o<strong>the</strong>r sources [12].9.2.2 Lymph Node Regions<strong>of</strong> <strong>the</strong> <strong>Neck</strong>The cervical lymph nodes can be divided into superficial<strong>and</strong> deep nodes, <strong>and</strong> each <strong>of</strong> <strong>the</strong>se groups into lateral<strong>and</strong> medial. The deep lateral nodes are distributedamong several large groups:1. The submental <strong>and</strong> subm<strong>and</strong>ibular group;2. The internal jugular chain (superior, middle, <strong>and</strong> inferior);3. The spinal accessory nerves chain;4. The supraclavicular node chain.Fig. 9.1. Cervical nodes by levels <strong>and</strong> sublevels: IA submental,IB subm<strong>and</strong>ibular, II upper jugular, IIA jugulogastric, IIB supraspinalaccessory, III middle jugular, IV lower jugular, V Posteriorcervical, VA spinal accessory nerve nodes, VB transverse cervicalnodes, SC supraclavicular, VI anterior groupThe deep medial cervical group consists <strong>of</strong> <strong>the</strong> prelaryngeal,prethyroidal, pretracheal <strong>and</strong> paratracheal lymphnodes. The superficial medial lymph nodes are distributedaround <strong>the</strong> anterior jugular vein. The superficiallateral cervical nodes are located along <strong>the</strong> externaljugular vein.Figure 9. 1 shows <strong>the</strong> system for describing <strong>the</strong> location<strong>of</strong> lymph nodes in <strong>the</strong> neck, <strong>and</strong> used <strong>the</strong> levels recommendedby <strong>the</strong> Committee for <strong>Head</strong> <strong>and</strong> <strong>Neck</strong> Surgery<strong>and</strong> Oncology <strong>of</strong> <strong>the</strong> American Academy for Otolaryngology-<strong>Head</strong><strong>and</strong> <strong>Neck</strong> Surgery [99].9.3 Cysts<strong>of</strong> <strong>the</strong> <strong>Neck</strong>Cysts <strong>of</strong> <strong>the</strong> neck are pathological cavities lined wi<strong>the</strong>pi<strong>the</strong>lium. The type <strong>of</strong> epi<strong>the</strong>lium varies, <strong>and</strong> <strong>the</strong> cavitymay contain fluid, keratin, mucus or o<strong>the</strong>r products.Cervical cysts can be divided into two large groups: developmental<strong>and</strong> non-developmental. Establishing <strong>the</strong>precise nature <strong>of</strong> <strong>the</strong>se cysts is important because <strong>the</strong>reare considerable differences in <strong>the</strong>ir biological <strong>and</strong> clini-


<strong>Neck</strong> Cysts, Metastasis, Dissection Chapter 9 265Table 9.1. Order <strong>of</strong> frequency <strong>of</strong> cervical cystic tumours according to age (extracted from [52, 68, 70, 115]). CA carcinomaInfants <strong>and</strong> children Adolescents AdultsThyroglossal duct cystBranchial cleft cystLymphangiomaHaemangiomaTeratoma <strong>and</strong> dermoidBronchogenic cystThymic cystLaryngoceleMetastatic thyroid caThyroglossal duct cystBranchial cleft cystBronchogenic cystThymic cystTeratoma <strong>and</strong> dermoidMetastatic thyroid caMetastatic cystic caThyroglossal duct cystCervical ranulaBranchial cleft cystLaryngoceleParathyroid cystThymic cystcal behaviour [26]. Because <strong>of</strong> <strong>the</strong> frequent similarities in<strong>the</strong> morphological aspects <strong>of</strong> various cysts, a definitivediagnosis is dependent on clinical data. These include <strong>the</strong>exact location <strong>of</strong> <strong>the</strong> lesion <strong>and</strong> <strong>the</strong> age <strong>of</strong> <strong>the</strong> patient. Theclinical manifestations <strong>of</strong> cysts depend largely on <strong>the</strong>irsize. Most cysts in <strong>the</strong> early stages are asymptomatic <strong>and</strong>are found on routine physical or radiographic examination.Rupture <strong>and</strong> drainage leads to infection, abscess,<strong>and</strong> sinus formation, which are frequently accompaniedby pain <strong>and</strong> swelling. In certain instances, computed tomographyscan can be <strong>of</strong> benefit in establishing <strong>the</strong> diagnosis<strong>and</strong>/or extension into adjacent structures [59].Aspiration needle biopsy can also be useful in distinguishingbetween cysts <strong>and</strong> o<strong>the</strong>r pathoses that presenta similar roentgenographic appearance [41].In adults, an asymptomatic neck mass should be consideredmalignancy until proven o<strong>the</strong>rwise. With <strong>the</strong> exception<strong>of</strong> thyroid nodules <strong>and</strong> salivary gl<strong>and</strong> tumours,neck masses in adults have <strong>the</strong> following characteristics:80% <strong>of</strong> <strong>the</strong> masses are neoplastic, 80% <strong>of</strong> neoplasticmasses are malignant, 80% <strong>of</strong> malignancies are metastatic,<strong>and</strong> in 80%, <strong>the</strong> primary tumour is located above<strong>the</strong> level <strong>of</strong> clavicle [70]. In contrast, 90% <strong>of</strong> neck massesin children represent benign conditions. In a review <strong>of</strong>445 children with neck masses, 55% <strong>of</strong> <strong>the</strong> masses werecongenital cysts, 27% were inflammatory lesions, 11%were malignant <strong>and</strong> 7% were miscellaneous conditions[117]. Table 9.1 lists <strong>the</strong> causes <strong>of</strong> neck masses in order<strong>of</strong> <strong>the</strong> frequency with which <strong>the</strong>y occur, according to <strong>the</strong>age <strong>of</strong> <strong>the</strong> patient.9.3.1 Developmental Cysts9.3.1.1 Branchial Cleft Cysts,Sinuses <strong>and</strong> FistulaeBranchial apparatus anomalies are lateral cervical lesionsthat result from congenital developmental defectsarising from <strong>the</strong> primitive branchial arches, clefts <strong>and</strong>pouches.The branchial apparatus appears around <strong>the</strong> 4thweek <strong>of</strong> gestation <strong>and</strong> gives rise to multiple structures orderivatives <strong>of</strong> <strong>the</strong> ears, face, oral cavity <strong>and</strong> neck. Thesestructures are described in more detail in o<strong>the</strong>r sources[126]. Anatomically, <strong>the</strong> branchial apparatus consists<strong>of</strong> a paired series <strong>of</strong> six arches, five internal pouches<strong>and</strong> five external clefts or grooves. The external groovesare <strong>of</strong> ectodermal origin <strong>and</strong> are called branchial clefts.The internal pouches are <strong>of</strong> endodermal origin <strong>and</strong> areknown as pharyngeal pouches; <strong>the</strong>y are separated by<strong>the</strong>ir branchial plates [126]. Each branchial arch is suppliedby an artery <strong>and</strong> a nerve <strong>and</strong> develops into well-definedmuscles, bone <strong>and</strong> cartilage. Thus, all three germcelllayers contribute to formation <strong>of</strong> <strong>the</strong> branchial apparatus.The arches are numbered 1–6, from cranial tocaudal, <strong>and</strong> <strong>the</strong> clefts <strong>and</strong> pouches 1–5. The correspondingcleft <strong>and</strong> pouch lie immediately caudal to <strong>the</strong>ir numericalarch, that is, <strong>the</strong> first cleft <strong>and</strong> pouch lie between<strong>the</strong> first <strong>and</strong> second arches, <strong>the</strong> second cleft <strong>and</strong> pouchlie between <strong>the</strong> second <strong>and</strong> third arches, <strong>and</strong> so on.A number <strong>of</strong> <strong>the</strong>ories exist to explain <strong>the</strong> genesis <strong>of</strong>branchial cleft anomalies. Regauer <strong>and</strong> associates haveproposed that <strong>the</strong> cysts arise from <strong>the</strong> endodermally derivedsecond branchial pouch [95]. An alternative explanationis that <strong>the</strong> cysts develop from cystic epi<strong>the</strong>lial inclusionsin lymph nodes that are ei<strong>the</strong>r <strong>of</strong> salivary gl<strong>and</strong>origin or from displaced epi<strong>the</strong>lium from <strong>the</strong> palatinetonsil [43]. Golledge <strong>and</strong> Ellis recently reviewed <strong>the</strong> various<strong>the</strong>ories on <strong>the</strong> histogenesis <strong>of</strong> branchial cleft cysts[43].Papers dealing with anomalies <strong>of</strong> <strong>the</strong> branchial apparatusdo not always distinguish between <strong>the</strong> terms sinus<strong>and</strong> fistula <strong>and</strong> <strong>of</strong>ten use <strong>the</strong>m interchangeably as synonyms.A sinus is a tract that has only one opening, ei<strong>the</strong>rcutaneous or mucosal. A fistula is a tract that hastwo openings, one cutaneous <strong>and</strong> one mucosal. A cystmay occur independently or in association with a sinusor fistula.Most anomalies <strong>of</strong> <strong>the</strong> branchial apparatus <strong>of</strong> concernto <strong>the</strong> surgical pathologist present clinically as acyst, fistula, sinus or skin tag. Fistulae, sinuses <strong>and</strong> skintags occur in younger patients than cysts do [20]. Bran-


266 M. A Luna · K. Pineda-Daboin9Fig. 9.2. Branchial cleft cyst. Wall lined with stratified squamousepi<strong>the</strong>liumchial cleft cysts constitute approximately 75–80% <strong>of</strong> allbranchial anomalies, <strong>and</strong> fistulae <strong>and</strong> sinuses toge<strong>the</strong>raccount for 15–20% <strong>of</strong> all such malformations [1]. Insome series, external fistulae, sinuses <strong>and</strong> skin tags aremore common than cysts [57].Of all branchial anomalies, 92–99% are associatedwith <strong>the</strong> second branchial clefts apparatus, probably becauseit is deeper <strong>and</strong> longer than <strong>the</strong> o<strong>the</strong>rs [8]. Use <strong>of</strong><strong>the</strong> name “branchial cyst” without fur<strong>the</strong>r qualificationgenerally refers to a cyst <strong>of</strong> second branchial origin. Cystsare three times more common than sinuses <strong>and</strong> fistulaein this apparatus. They typically occur along <strong>the</strong> anteriorborder <strong>of</strong> <strong>the</strong> sternocleidomastoid muscle from <strong>the</strong> hyoidbone to <strong>the</strong> suprasternal notch, but have infrequentlybeen described in <strong>the</strong> midline, just as a thyroglossal ductcyst may occur laterally, as bilateral branchial cleft cysts,or even in <strong>the</strong> lateral wall <strong>of</strong> <strong>the</strong> nasopharynx [8, 85].Branchial cleft anomalies have no gender preference.Most patients (75%) are aged 20–40 years at <strong>the</strong> time <strong>of</strong>diagnosis. Since fewer than 3% <strong>of</strong> cysts are found in patientsolder than 50 years, <strong>the</strong> pathologist must be carefulin making this diagnosis in this age group; a metastaticcystic squamous cell carcinoma in a cervicallymph node may masquerade as a branchial cleft cyst.On pathologic examination <strong>the</strong> cysts are unilocular,usually between 2 <strong>and</strong> 6 cm in diameter, <strong>and</strong> lined withstratified squamous epi<strong>the</strong>lium (90%), respiratory epi<strong>the</strong>lium(8%), or both (2%; Fig. 9.2). Lymphoid aggregateswith or without reactive germinal centres beneath<strong>the</strong> lining epi<strong>the</strong>lium are found in <strong>the</strong> majority <strong>of</strong> cysts(75–80%). Acute <strong>and</strong> chronic inflammation, foreignbody giant-cell reaction <strong>and</strong> fibrosis are <strong>the</strong> secondarymicroscopic changes in <strong>the</strong> wall <strong>of</strong> <strong>the</strong> cyst. In exceptionalcases, heterotopic salivary tissue may even befound in <strong>the</strong> wall <strong>of</strong> <strong>the</strong> cyst [111]. Carcinoma in situ hasseldom been described in <strong>the</strong> lining <strong>of</strong> <strong>the</strong> cysts [132].Regauer et al. have postulated that <strong>the</strong> cysts are initiallylined with <strong>the</strong> endodermally derived pouch type <strong>of</strong> respiratoryepi<strong>the</strong>lium, which is replaced by squamous epi<strong>the</strong>liumthrough an intermediate stage <strong>of</strong> pseudostratifiedtransitional-type epi<strong>the</strong>lium [95].Fistulae <strong>and</strong> sinuses are more <strong>of</strong>ten found at birthor in early childhood than cysts. The external opening,when present, is usually located along <strong>the</strong> anterior border<strong>of</strong> <strong>the</strong> sternocleidomastoid muscle at <strong>the</strong> junction <strong>of</strong>its middle <strong>and</strong> lower thirds. The tract, if <strong>the</strong>re is one,follows <strong>the</strong> carotid sheath; it crosses over <strong>the</strong> hypoglossalnerve, runs between <strong>the</strong> internal <strong>and</strong> external carotidarteries <strong>and</strong> ends at <strong>the</strong> tonsillar fossa [122].Thymic cyst <strong>and</strong> cystic low-grade mucoepidermoidcarcinoma with prominent lymphoid stroma are considerationsin <strong>the</strong> differential diagnosis. The cyst’s benignlining distinguishes it from metastatic cystic squamouscarcinoma.Complete surgical excision <strong>of</strong> <strong>the</strong> cyst, sinus, or fistulais indicated. In a review <strong>of</strong> 274 patients with branchialremnants treated at <strong>the</strong> Mayo Clinic, <strong>the</strong> recurrence ratewas only 2.7% for patients with no history <strong>of</strong> surgery orinfection, 14% in those with a history <strong>of</strong> infection <strong>and</strong>21.2% in those who had undergone prior attempts at surgicalremoval [28].Anomalies from <strong>the</strong> first branchial arch accountedfor only 8% <strong>of</strong> all branchial cleft anomalies at <strong>the</strong> MayoClinic [82]. Of <strong>the</strong>se, 68% were cysts, 16% sinuses <strong>and</strong>16% fistulae. These anomalies occur predominantly infemales <strong>and</strong> are found in all age groups. In general, sinuses<strong>and</strong> fistulae tend to develop in infants <strong>and</strong> children,whereas cysts are more common in older groups.Clinically, <strong>the</strong>y may masquerade as parotid tumours oras otitis with ear drainage [82].Disorders <strong>of</strong> <strong>the</strong> first branchial cleft are classified intotwo types [119]. Type I are those that embryologically duplicate<strong>the</strong> membrane (cutaneous) external auditory canal.Accordingly, only ectodermal components are observedunder <strong>the</strong> microscope. On histologic examination<strong>the</strong>y are <strong>of</strong>ten confused with epidermoid cysts, for <strong>the</strong>yare lined solely by keratinised, stratified squamous epi<strong>the</strong>lium,with no adnexal structures or cartilage. Characteristically,<strong>the</strong>y are located medial, inferior or posteriorto <strong>the</strong> concha <strong>and</strong> pinna. Drainage from cysts or fistulaemay occur in any <strong>of</strong> <strong>the</strong>se sites. The fistula tract or sinusmay parallel <strong>the</strong> external auditory canal <strong>and</strong> ends in ablind cul de sac at <strong>the</strong> level <strong>of</strong> <strong>the</strong> mesotympanum.Type II deformities are composed <strong>of</strong> both ectodermal<strong>and</strong> mesodermal elements <strong>and</strong> <strong>the</strong>refore contain,in addition to skin, cutaneous appendages <strong>and</strong> cartilage(Fig. 9.3). Patients with this defect usually present withan abscess or fistula at a point just below <strong>the</strong> angle <strong>of</strong> <strong>the</strong>m<strong>and</strong>ible, through <strong>the</strong> parotid gl<strong>and</strong>, toward <strong>the</strong> externalauditory canal. Type II defects are <strong>the</strong>refore more intimatelyassociated with <strong>the</strong> parotid gl<strong>and</strong> than are type Idefects. Sometimes an anomaly cannot be distinguishedas type I or type II. In those instances, Olsen et al. suggestedthat <strong>the</strong> abnormality be classified only as to whe<strong>the</strong>rit is a cyst, sinus, or fistula [82]. Complete excision is


<strong>Neck</strong> Cysts, Metastasis, Dissection Chapter 9 267Fig. 9.3. First branchial cleft cyst, Type II. Squamous epi<strong>the</strong>liumlining <strong>the</strong> cystic cavity. Note <strong>the</strong> presence <strong>of</strong> skin appendage structures<strong>and</strong> cartilage in <strong>the</strong> stromaFig. 9.5. Branchogenic carcinoma. Inset: malignant squamous<strong>and</strong> respiratory epi<strong>the</strong>lium lining <strong>the</strong> cystic wallNei<strong>the</strong>r <strong>the</strong> fifth nor <strong>the</strong> sixth branchial arch formsclefts or pouches in humans [126]. Branchial cleft cystshave been reported infrequently in <strong>the</strong> parotid, thyroid<strong>and</strong> parathyroid gl<strong>and</strong>s, floor <strong>of</strong> <strong>the</strong> mouth, tonsil,pharynx <strong>and</strong> mediastinum [17]. Many <strong>of</strong> <strong>the</strong>se cystshave <strong>the</strong> microscopic features <strong>of</strong> lymphoepi<strong>the</strong>lial cysts(Fig. 9.4).9.3.2 Branchiogenic CarcinomaFig. 9.4. Lymphoepi<strong>the</strong>lial cyst. Note <strong>the</strong> absence <strong>of</strong> lymphoidhyperplasia<strong>the</strong> only effective treatment. In some cases, this may necessitatea superficial parotidectomy. First branchial cleftabnormalities must be differentiated pathologically fromepidermal cysts (especially type I), dermoids (especiallytype II), <strong>and</strong> cystic sebaceous lymphadenoma.Anomalies from <strong>the</strong> third <strong>and</strong> fourth branchial apparatusesare rare <strong>and</strong> toge<strong>the</strong>r account for fewer than 5% <strong>of</strong>all branchial cysts, sinuses <strong>and</strong> fistulae [80]. A fistula in<strong>the</strong> pyriform sinus is one <strong>of</strong> <strong>the</strong> more common manifestations<strong>of</strong> a third branchial anomaly [32]. Recurrent infections<strong>of</strong> <strong>the</strong> lower neck, including suppurative thyroiditis,<strong>and</strong> a fistulous tract into <strong>the</strong> pyriform sinus are <strong>the</strong> features<strong>of</strong> a fourth branchial cleft or pouch anomaly [80].Third <strong>and</strong> fourth branchial sinus anomalies can bedistinguished only by detailed surgical exploration. Athird branchial sinus always extends from <strong>the</strong> pyriformsinus through <strong>the</strong> thyroid membrane cranial to <strong>the</strong> superiorlaryngeal nerve. In contrast, a fourth branchialsinus extends from <strong>the</strong> pyriform sinus caudal to <strong>the</strong> superiorlaryngeal nerve <strong>and</strong> exits <strong>the</strong> larynx near <strong>the</strong> cricothyroidjoint [127].Branchiogenic carcinoma or primary cervical neoplasticcysts are <strong>of</strong> interest from an historical viewpoint [69,96]. Few <strong>of</strong> <strong>the</strong> purported examples <strong>of</strong> this entity fulfil<strong>the</strong> four criteria that Martin et al. considered necessaryto establish <strong>the</strong> diagnosis, which are as follows:1. The cervical tumour occurs along <strong>the</strong> line extendingfrom a point just anterior to <strong>the</strong> tragus, along <strong>the</strong>anterior border <strong>of</strong> <strong>the</strong> sternocleidomastoid muscle, to<strong>the</strong> clavicle;2. The histologic appearance must be consistent with anorigin from tissue known to be present in <strong>the</strong> branchialvestigial;3. No primary source <strong>of</strong> <strong>the</strong> carcinoma should be discoveredduring follow-up for at least 5 years;4. Cancer arising in <strong>the</strong> wall <strong>of</strong> an epi<strong>the</strong>lium-lined cystsituated in <strong>the</strong> lateral aspect <strong>of</strong> <strong>the</strong> neck can be demonstratedhistologically (Fig. 9.5) [69].The fulfilment <strong>of</strong> <strong>the</strong>se criteria is practically impossible,<strong>and</strong> <strong>the</strong> existence <strong>of</strong> “ branchiogenic carcinoma” mustremain entirely hypo<strong>the</strong>tical [69, 73, 96, 116]. The criteriahave been criticised on <strong>the</strong> grounds that <strong>the</strong>y aremuch too restrictive <strong>and</strong> nearly preclude a diagnosis <strong>of</strong>branchiogenic carcinoma [15, 86].Several authors have estimated that, even acceptingtentative examples <strong>of</strong> branchiogenic carcinoma, its incidencewould be minuscule (0.3% <strong>of</strong> all malignant su-


268 M. A Luna · K. Pineda-Daboin9praclavicular tumours) [15, 58]. There is no doubt thatmost, if not all <strong>of</strong> <strong>the</strong>m are actually cervical node metastaseswith a cystic pattern. The palatine tonsil, or moregenerally <strong>the</strong> anatomic region <strong>of</strong> Waldeyer’s ring, is notoriousfor producing cystic solitary metastases that resemble<strong>the</strong> usual appearance <strong>of</strong> branchial cleft cysts [69,73, 96, 116].All <strong>of</strong> <strong>the</strong> suspected branchiogenic carcinomas havebeen squamous-cell in type, <strong>and</strong> all but one have beenin <strong>the</strong> region <strong>of</strong> <strong>the</strong> second branchial apparatus [15, 58,86]. The patients have been predominantly males rangingin age from 38 to 71 years [15, 58]. Nearly all <strong>of</strong> <strong>the</strong>semasses have been cystic <strong>and</strong> have resided in a lymphoidmatrix, hence <strong>the</strong> presumed relationship to a branchialcyst. It should be obvious that nei<strong>the</strong>r cystic architecturenor association with lymphoid tissue is, in itself, an acceptablecriterion for diagnosis <strong>of</strong> branchiogenic carcinoma[73, 96, 116].An absence or presence or peripheral lymphatic sinuses<strong>and</strong>/or follicular centres in <strong>the</strong> lymphoid tissuehas been used to exclude or confirm metastasis to <strong>the</strong>lymph nodes. This criterion is not valid. Branchial cleftcysts <strong>of</strong>ten lie within lymph nodes, <strong>and</strong> metastases canobscure <strong>the</strong> architecture <strong>of</strong> a lymph node [96, 116].9.3.3 Thyroglossal Duct Cyst<strong>and</strong> Ectopic ThyroidCysts <strong>and</strong> sinuses may be found along <strong>the</strong> course <strong>of</strong> <strong>the</strong>thyroglossal duct; <strong>the</strong>se cysts develop during <strong>the</strong> migration<strong>of</strong> <strong>the</strong> thyroid gl<strong>and</strong> from <strong>the</strong> base <strong>of</strong> <strong>the</strong> tongue.The cysts are situated in <strong>the</strong> midline <strong>of</strong> <strong>the</strong> neck, usuallybelow <strong>the</strong> hyoid bone. A fistula may develop froman infected cyst.The thyroid begins to develop during <strong>the</strong> 4th week<strong>of</strong> gestation when <strong>the</strong> embryo is about 2–2.5 mm long[126]. It is an endodermal derivative composed <strong>of</strong> twosmall lateral anlagen <strong>and</strong> <strong>the</strong> more substantial mediananlage from <strong>the</strong> foramen caecum at <strong>the</strong> base <strong>of</strong><strong>the</strong> tongue. Because <strong>of</strong> elongated cephalad embryonicgrowth ra<strong>the</strong>r than active descent, <strong>the</strong> orthotopic pretracheallocation <strong>of</strong> <strong>the</strong> thyroid is caudal to <strong>the</strong> foramencaecum [126].Thyroglossal duct cysts (TDC) are twice as commonas branchial cleft cysts. In a review <strong>of</strong> 1,534 cases in <strong>the</strong>literature, Allard observed that, at <strong>the</strong> time <strong>of</strong> presentation,67% <strong>of</strong> patients had a cyst <strong>and</strong> 33% a fistula [5].Approximately 90% <strong>of</strong> TDC occur in <strong>the</strong> midline <strong>of</strong><strong>the</strong> neck, although some may occur paramedially, most<strong>of</strong>ten on <strong>the</strong> left. Overall, 73.8% occur below <strong>the</strong> hyoidbone, 24.1% are suprahyoid, <strong>and</strong> 2.1% are intralingual[65]. Spinelli et al. reviewed <strong>the</strong>ir experience with neckmasses in children <strong>and</strong> noted that 17 (26%) <strong>of</strong> 154 caseswere TDC, <strong>and</strong> branchial cleft cysts were less common[115].Most patients with a TDC have no symptoms; <strong>the</strong>yseek evaluation for a midline neck mass discovered incidentallyby <strong>the</strong>mselves or a family member. The mostcommon manifestations are pain, a draining sinus orfistula, infection, or dysphagia. A cyst in <strong>the</strong> floor <strong>of</strong><strong>the</strong> mouth may cause feeding problems in newborns,whereas a cyst at <strong>the</strong> base <strong>of</strong> <strong>the</strong> tongue has, in rare instances,been responsible for sudden death in infancy[106].Thyroglossal duct cysts range in size from 0.5 to4 cm in diameter. They can be ei<strong>the</strong>r unilocular or multilocular<strong>and</strong> usually contain mucoid material if <strong>the</strong> cystis not infected or mucopurulent material or pus if it is.The type <strong>of</strong> epi<strong>the</strong>lium lining <strong>the</strong> cyst varies from onecase to ano<strong>the</strong>r, or even within <strong>the</strong> same surgical specimen.A columnar to stratified cuboidal epi<strong>the</strong>lium withcilia is <strong>the</strong> most common type <strong>of</strong> epi<strong>the</strong>lial lining, foundin 50–60% <strong>of</strong> cases (Fig. 9.6). Lymphoid nodules in <strong>the</strong>wall <strong>of</strong> <strong>the</strong> cyst are found in 15–20% <strong>of</strong> cases, while <strong>the</strong>yoccur in 75% <strong>of</strong> branchial cleft cysts. A TDC with squamouslining <strong>and</strong> lymphoid tissue may be difficult to differentiatefrom a branchial cleft cyst. Immunoperoxidasestaining for thyroglobulin may be <strong>of</strong> help. Ectopicthyroid tissue is identified (as collections <strong>of</strong> thyroid folliclesin <strong>the</strong> s<strong>of</strong>t tissues adjacent to <strong>the</strong> cyst) in 3 –20%<strong>of</strong> TDCs, although <strong>the</strong>se figures are related to some extentto <strong>the</strong> number <strong>of</strong> tissue slides taken for histologicexamination <strong>and</strong> <strong>the</strong> extent <strong>of</strong> inflammatory <strong>and</strong> reactivechanges present in <strong>the</strong> surrounding tissue.Mucous gl<strong>and</strong>s were identified in 60% <strong>of</strong> <strong>the</strong> TDCstudied by Sade <strong>and</strong> Rosen [102]. These authors believe<strong>the</strong> mucous gl<strong>and</strong>s to be part <strong>of</strong> <strong>the</strong> normal thyroglossalapparatus <strong>and</strong> not just gl<strong>and</strong>s found at <strong>the</strong> base <strong>of</strong><strong>the</strong> tongue.Thyroglossal duct remnants are treated by completesurgical excision using <strong>the</strong> Sistrunk operation [112].This consists <strong>of</strong> a block excision <strong>of</strong> <strong>the</strong> entire thyroglossaltract to <strong>the</strong> foramen caecum, as well as removal <strong>of</strong><strong>the</strong> central 1–2 cm <strong>of</strong> <strong>the</strong> hyoid bone. If this procedure isperformed, <strong>the</strong> TDC recurrence rate is less than 5% [91].If <strong>the</strong> central portion <strong>of</strong> <strong>the</strong> bone is not removed, <strong>the</strong> recurrencerate is as high as 50% [3, 74].Ectopic thyroid is defined by identification <strong>of</strong> gross ormicroscopic thyroid tissue outside <strong>of</strong> <strong>the</strong> thyroid gl<strong>and</strong>.Most commonly from <strong>the</strong> base <strong>of</strong> <strong>the</strong> tongue (lingualthyroid) to <strong>the</strong> mid-lower neck superior to <strong>the</strong> orthotopicthyroid [25]. The ectopia can be complete or more <strong>of</strong>tenassociated with an orthotopic thyroid.Hypothyroidism is a frequent finding in patients withlingual thyroid [105]. Batsakis <strong>and</strong> collaborators noted aclinical prevalence <strong>of</strong> lingual thyroid <strong>of</strong> 1 in 10,000 individuals,but an autopsy prevalence <strong>of</strong> 1 in 10 [10]. Ectopicthyroid is histologically composed <strong>of</strong> uniform, <strong>of</strong>tensmall, follicles containing minimal colloid. The micr<strong>of</strong>olliclesare usually intercepted by <strong>the</strong> skeletal muscle<strong>of</strong> <strong>the</strong> tongue.


<strong>Neck</strong> Cysts, Metastasis, Dissection Chapter 9 269Fig. 9.6. Thyroglossal duct cyst. Thyroid follicles are present in<strong>the</strong> wallFig. 9.7. Cervical thymic cyst. Notice Hassall’s corpuscles in <strong>the</strong>wall (arrow)Patients with presumed ectopic thyroid should undergoa preoperative thyroid scan to rule out ectopicthyroid gl<strong>and</strong>, because patients with an ectopic thyroidgl<strong>and</strong> have no additional normally functional tissue,<strong>and</strong> thus are rendered permanently athyroid by excision<strong>of</strong> <strong>the</strong> ectopic gl<strong>and</strong> [88].Carcinoma from <strong>the</strong> thyroglossal duct (Hürthle celladenomas <strong>and</strong> papillary carcinomas) have been reportedin less than 1% <strong>of</strong> TDC [6, 118]. Yoo et al. reviewed115 cases <strong>of</strong> papillary carcinoma arising in TDC publishedin <strong>the</strong> literature [128]. These tumours are typicallyintracystic, <strong>and</strong> usually <strong>the</strong> thyroid gl<strong>and</strong> proper isuninvolved. However, multifocal papillary carcinomashave been documented in TDC as well as in <strong>the</strong> gl<strong>and</strong> [6,118]. Most researchers agree that a) total thyroidectomyis not routinely indicated as long as <strong>the</strong>re are no palpableabnormalities in <strong>the</strong> gl<strong>and</strong> <strong>and</strong> no significant scintiscanfindings, <strong>and</strong> b) <strong>the</strong> Sistrunk operation probably <strong>of</strong>fers areasonable chance <strong>of</strong> cure [6, 63, 118, 128].9.3.4 Cervical Thymic CystFaulty development <strong>of</strong> <strong>the</strong> third <strong>and</strong> fourth pharyngealpouches results in abnormalities <strong>of</strong> <strong>the</strong> thymus <strong>and</strong>parathyroid gl<strong>and</strong>s.Cervical thymic cysts (CTC) are morphologicallyidentical to <strong>the</strong>ir mediastinal counterparts. They arefound in <strong>the</strong> anterior triangle <strong>of</strong> <strong>the</strong> neck along <strong>the</strong> normalpath <strong>of</strong> descent <strong>of</strong> <strong>the</strong> thymus, with or without parathyroidgl<strong>and</strong>s, <strong>and</strong> <strong>the</strong>y have a fibrous b<strong>and</strong> or a solidthymic cord connection to <strong>the</strong> pharynx or mediastinum.The thymus develops as paired structures from <strong>the</strong>third branchial pouch in <strong>the</strong> 6th week <strong>of</strong> gestation.The endodermal primordium <strong>of</strong> <strong>the</strong> thymus has a ductalor luminal connection to <strong>the</strong> pouch that is knownas <strong>the</strong> thymopharyngeal duct. Ventromedial <strong>and</strong> caudalgrowth <strong>of</strong> <strong>the</strong> respective anlage results in separation<strong>of</strong> <strong>the</strong> thymus from <strong>the</strong> pharynx. The fragmented remnants<strong>of</strong> <strong>the</strong> solid thymopharyngeal duct are thought tobe <strong>the</strong> progenitors <strong>of</strong> accessory parathyroid <strong>and</strong> thymictissue in <strong>the</strong> neck [62]. The inferior parathyroid gl<strong>and</strong>salso originate from <strong>the</strong> third pouch, <strong>and</strong> <strong>the</strong>ir descentwith <strong>the</strong> thymus explains <strong>the</strong>ir localisation relative to<strong>the</strong> superior parathyroids, which arise from <strong>the</strong> fourthbranchial pouch. By <strong>the</strong> end <strong>of</strong> <strong>the</strong> 8th week, <strong>the</strong> lowerpoles <strong>of</strong> <strong>the</strong> thymic anlage approach each o<strong>the</strong>r, but donot fuse, at <strong>the</strong> level <strong>of</strong> <strong>the</strong> aortic arch. Failures to involuteor descend <strong>of</strong> any <strong>of</strong> <strong>the</strong> thymic anlage are responsiblefor a variety <strong>of</strong> abnormalities, such as thymic cysts.The reader is referred to <strong>the</strong> excellent paper by Zarbo etal. for <strong>the</strong> classification <strong>of</strong> <strong>the</strong>se developmental abnormalities<strong>of</strong> <strong>the</strong> thymus [130].Cervical thymic cysts are uncommon; approximately120 cases in children were reported through 2001 [51,89]. Males are affected more commonly than females. Accordingto Guba et al., 70% <strong>of</strong> CTC are on <strong>the</strong> left side <strong>of</strong><strong>the</strong> neck, 23% on <strong>the</strong> right <strong>and</strong> <strong>the</strong> remainder in <strong>the</strong> midline[47]. They can be found anywhere from <strong>the</strong> angle <strong>of</strong><strong>the</strong> m<strong>and</strong>ible to <strong>the</strong> sternum, paralleling <strong>the</strong> sternocleidomastoidmuscle <strong>and</strong> normal descent <strong>of</strong> <strong>the</strong> thymus. Sixtysevenpercent occur in <strong>the</strong> first decade <strong>of</strong> life. The remainderoccur in <strong>the</strong> second <strong>and</strong> third decades [51].The cysts range between 2 <strong>and</strong> 15 cm <strong>and</strong> may be ei<strong>the</strong>runilocular or multilocular. The epi<strong>the</strong>lial liningmay be cuboidal, columnar, or stratified squamous. Insome areas, <strong>the</strong> epi<strong>the</strong>lium may be replaced by fibrous orgranulation tissue containing cholesterol clefts <strong>and</strong> multinucleatedgiant cells. To qualify a cyst as a CTC, thymictissue must be found within <strong>the</strong> cyst wall (Fig. 9.7);detection <strong>of</strong> this tissue may require numerous sections.Cervical thymic cysts rarely have malignant potential.Recently, Moran et al. reported for <strong>the</strong> first timecarcinomas arising in CTC [76]. This contrasts with mediastinalthymic cysts, in which malignancies are <strong>of</strong>tenseen. CTC also have not demonstrated pseudoepi<strong>the</strong>liomatoushyperplastic changes, as in some mediastinalcysts.


270 M. A Luna · K. Pineda-DaboinFig. 9.8. Cervical parathyroid cyst lined with cuboidal epi<strong>the</strong>lium.Parathyroid tissue is present in <strong>the</strong> wallFig. 9.9. Cervical bronchogenic cyst. Respiratory epi<strong>the</strong>lium lines<strong>the</strong> cyst wall9The differential diagnosis includes <strong>the</strong> o<strong>the</strong>r developmentalcysts <strong>of</strong> <strong>the</strong> neck, as well as rare cystic presentations<strong>of</strong> Hodgkin’s disease, thymoma, <strong>and</strong> germinoma.These three neoplasms are more likely, however, to presentin <strong>the</strong> anterior mediastinum. The reader is referredto <strong>the</strong> excellent paper by May for clinical features usefulin differentiating <strong>the</strong>se conditions [70]. Complete surgicalexcision is <strong>the</strong> treatment <strong>of</strong> choice.9.3.5 Cervical ParathyroidCystCysts <strong>of</strong> <strong>the</strong> parathyroid gl<strong>and</strong>s, like thymic cysts, haveseveral characteristic morphologic features, including apersistent hollow tract with <strong>the</strong> third or fourth branchialpouch. It is estimated that 5% <strong>of</strong> neck cysts or fewer areparathyroid in origin. Few parathyroid cysts have beenreported in childhood, implying that most are acquired,including cystic parathyroid adenoma.There appear to be two distinct types <strong>of</strong> parathyroidcysts: non-functioning <strong>and</strong> functioning. The formermake up <strong>the</strong> majority <strong>of</strong> <strong>the</strong>se cysts <strong>and</strong> are about twoto three times more common in women than in men.The mean age <strong>of</strong> patients with a non-functioning cyst is43.3 years. Functioning cysts account for 11.5 to 30% <strong>of</strong><strong>the</strong>se cysts [48]. They are more common in men by a ratio<strong>of</strong> 1.6:1 <strong>and</strong> tend to occur in sites o<strong>the</strong>r than <strong>the</strong> inferiorparathyroid gl<strong>and</strong>s, from <strong>the</strong> angle <strong>of</strong> <strong>the</strong> m<strong>and</strong>ibleto <strong>the</strong> mediastinum [120]. The mean age <strong>of</strong> patients withfunctioning cysts is 51.9 years.About 95% <strong>of</strong> <strong>the</strong>se cysts occur below <strong>the</strong> inferiorthyroid border, <strong>and</strong> 65% are associated with <strong>the</strong> inferiorparathyroid gl<strong>and</strong>s. Cysts have been identified from<strong>the</strong> angle <strong>of</strong> <strong>the</strong> m<strong>and</strong>ible to <strong>the</strong> mediastinum, however,<strong>and</strong> <strong>the</strong>y can occur in <strong>the</strong> thyroid lobe or posteriorly[36].Fine-needle aspiration is <strong>the</strong> principal diagnostictool. Aspiration <strong>of</strong> clear fluid with an elevated parathyroidhormone level is a definite indication <strong>of</strong> a parathyroidcyst. The C-terminal/midmolecular zone <strong>of</strong> <strong>the</strong>parathyroid hormone should be assayed, because <strong>the</strong>N-terminal-specific assay is frequently associated withfalse-negative results [81].Histologic studies show that a parathyroid cyst’s wallis usually formed by a solitary layer <strong>of</strong> compressed cuboidalor low columnar epi<strong>the</strong>lium, with ei<strong>the</strong>r chief oroxyphilic cells present in <strong>the</strong> fibrous capsule (Fig. 9.8).Some cysts may not have any identifiable parathyroidtissue, but even in <strong>the</strong>se cases a diagnosis can be establishedby testing <strong>the</strong> cystic fluid. Immunostaining forparathyroid hormone could be <strong>of</strong> help.Aspiration may be curative, but persistence or recurrence<strong>of</strong> <strong>the</strong> cyst is a sign that surgical removal is in order.Functional cysts are associated with a high risk <strong>of</strong>o<strong>the</strong>r parathyroid gl<strong>and</strong> abnormalities such as hyperplasiaor adenoma [92].9.3.6 Cervical BronchogenicCystCervical bronchogenic cysts are uncommon congenitallesions found almost invariably in <strong>the</strong> skin or subcutaneoustissue in <strong>the</strong> vicinity <strong>of</strong> <strong>the</strong> suprasternal notch or manubriumsterni, rarely in <strong>the</strong> anterior neck or shoulder.Bronchial cysts are derived from small buds <strong>of</strong> diverticulathat separate from <strong>the</strong> foregut during formation <strong>of</strong><strong>the</strong> tracheobronchial tree. When <strong>the</strong>y occur outside <strong>the</strong>thoracic cavity, <strong>the</strong> cyst presumably arises from erraticmigration <strong>of</strong> sequestered primordial cells.They are usually discovered at or soon after birth<strong>and</strong> appear as asymptomatic nodules that slowly increasein size or as draining sinuses exuding a mucoidmaterial. They are more common in males, in some seriesby a margin <strong>of</strong> 3:1 [31]. The cysts range from 0.3to 6 cm in size. They are lined by ciliated, pseudostratifiedcolumnar epi<strong>the</strong>lium (Fig. 9.9). If <strong>the</strong> cyst is infect-


<strong>Neck</strong> Cysts, Metastasis, Dissection Chapter 9 271ed squamous epi<strong>the</strong>lium is found. The cyst wall containssmooth muscle, elastic fibres <strong>and</strong> seromucous gl<strong>and</strong>s. In<strong>the</strong> 30 cases studied by Fraga et al., smooth muscle wasidentified in 24 <strong>and</strong> seromucous gl<strong>and</strong>s in 16. In contrastto <strong>the</strong>ir intrathoracic counterparts, only 2 containedcartilage [39].A bronchogenic cyst can be distinguished from a teratomaby a complete absence <strong>of</strong> tissues o<strong>the</strong>r than thosethat can be explained on <strong>the</strong> basis <strong>of</strong> a malformation.The lack <strong>of</strong> ciliated epi<strong>the</strong>lium distinguishes a lateralcervical cyst containing gastric mucosa from a cervicalbronchogenic cyst. TDC can be differentiated from abronchogenic cyst by finding thyroid follicles; fur<strong>the</strong>rmore,TDC do not contain smooth muscle or cartilage.Complete surgical excision <strong>of</strong> a bronchogenic cystalong with its sinus tract is curative. Malignancies havenot been described in cervical bronchogenic cysts.9.3.7 Dermoid CystICD-O:9084/0The term dermoid cyst should be reserved for a cysticneoplasm that originates from <strong>the</strong> ectoderm <strong>and</strong> mesoderm;endoderm is never found in <strong>the</strong>se cysts [101]. Thehead <strong>and</strong> neck area is a common site <strong>of</strong> occurrence fordermoid cysts, accounting for 34% <strong>of</strong> cases. These cystsare located in <strong>the</strong> skin <strong>and</strong> subcutaneous tissues [113].The position <strong>of</strong> <strong>the</strong>se dermoid cysts at <strong>the</strong> midline<strong>and</strong> along <strong>the</strong> lines <strong>of</strong> embryonic fusion <strong>of</strong> <strong>the</strong> facialprocesses is consistent with <strong>the</strong>ir origin by inclusions <strong>of</strong>ectodermal tissue along lines <strong>of</strong> closure at junctions <strong>of</strong>bone, s<strong>of</strong>t tissue, <strong>and</strong> embryonic membranes [87].Dermoid cysts in <strong>the</strong> neck account for 22% <strong>of</strong> midlineor near-midline neck lesions [101]. They have beendescribed in <strong>the</strong> upper neck, near <strong>the</strong> thyroid cartilage,<strong>and</strong> as low as <strong>the</strong> suprasternal notch. They may occur inpeople <strong>of</strong> almost any age. More than 50% are detected by<strong>the</strong> time a person is 6 years old, <strong>and</strong> approximately onethirdare present at birth [87, 101, 113]. The distributionbetween <strong>the</strong> sexes is approximately equal.Dermoid cysts range in size from a few millimetresto 12 cm in diameter. On microscopic examination <strong>the</strong>yare lined by stratified squamous epi<strong>the</strong>lium supportedby a fibrous connective tissue wall. Ectodermal derivativesmay be seen, including dermal adnexa such as hairfollicles, sebaceous gl<strong>and</strong>s, <strong>and</strong> sweat gl<strong>and</strong>s.9.3.8 Unclassified CervicalCystSome cysts may be difficult to classify because <strong>of</strong> anapparent discrepancy between <strong>the</strong> anatomic site <strong>of</strong> presentation<strong>and</strong> <strong>the</strong> histologic features, indeterminate microscopicfindings, loss <strong>of</strong> an intact epi<strong>the</strong>lial lining, ormixed histologic appearance. When a final determinationregarding <strong>the</strong> type <strong>of</strong> cyst is not possible, <strong>the</strong> term“congenital or developmental cyst, indeterminate type”should be used [121].9.3.9 Non-DevelopmentalCystsMost <strong>of</strong> <strong>the</strong> non-developmental cysts in <strong>the</strong> head <strong>and</strong>neck region occur in <strong>the</strong> jaw bones, oral cavity or parenchymaorgans, such as <strong>the</strong> thyroid gl<strong>and</strong>, salivarygl<strong>and</strong>s <strong>and</strong> parathyroid gl<strong>and</strong>s. Mucoceles, ranulas, <strong>and</strong>laryngoceles are considered non-developmental cyststhat may occur in <strong>the</strong> neck. Ranulas are actually pseudocysts:<strong>the</strong>y lack an epi<strong>the</strong>lial lining. Because <strong>the</strong>y mimictrue cysts histopathologically as well as clinically or radiographically,however, it is reasonable <strong>and</strong> convenientto include <strong>the</strong>m in a general discussion <strong>of</strong> cystic lesions.Mucoceles are discussed in Chap. 5.9.3.9.1 RanulaA ranula is a special type <strong>of</strong> mucous retention cyst mostcommonly caused by partial obstruction <strong>of</strong> <strong>the</strong> excretoryduct <strong>of</strong> <strong>the</strong> sublingual gl<strong>and</strong>. Rarely, it may originatefrom <strong>the</strong> cervical sinus or from branchial cleft remnants.The classic or simple ranula is a true cyst linedwith cuboidal, columnar or squamous epi<strong>the</strong>lium <strong>and</strong>filled with mucoid material similar to that found in mucoceles.It produces a mass in <strong>the</strong> floor <strong>of</strong> <strong>the</strong> mouth toone side <strong>of</strong> <strong>the</strong> midline. Dissection <strong>of</strong> <strong>the</strong> mucus into <strong>the</strong>fascial planes <strong>of</strong> <strong>the</strong> neck results in a pseudocyst calledplunging ranula. Simple ranulas are distinguished frommucoceles by <strong>the</strong>ir location <strong>and</strong> by <strong>the</strong> presence <strong>of</strong> anepi<strong>the</strong>lial lining [11].The plunging ranula may mimic o<strong>the</strong>r cystic or gl<strong>and</strong>ularswellings, such as dermoid <strong>and</strong> epidermoid cysts,TDC or cystic hygroma. Quick <strong>and</strong> Lowell [90] havepointed out that no specific clinical diagnostic tests areavailable to distinguish <strong>the</strong>se lesions. Consequently, adefinitive diagnosis is dependent on postoperative histopathologicevaluation <strong>of</strong> <strong>the</strong> surgical specimen.The management <strong>of</strong> <strong>the</strong>se lesions requires removal <strong>of</strong><strong>the</strong> sublingual gl<strong>and</strong> <strong>and</strong> excision <strong>of</strong> <strong>the</strong> ranula. The recurrencerate after this procedure is 0%. Excision <strong>of</strong> only<strong>the</strong> ranula was followed by a 25% recurrence rate, whereasmarsupialisation results in a 36% recurrence rate [129].9.3.9.2 LaryngoceleLaryngocele is a dilatation <strong>of</strong> Morgagni’s ventricle or itsappendages, which is filled with air or fluid [16, 37, 49].For more details on this lesion, see Chap. 7.


272 M. A Luna · K. Pineda-Daboin9.4 Cystic Neoplasms9.4.1 Cystic Hygroma<strong>and</strong> Lymphangioma9Cystic hygroma <strong>and</strong> lymphangioma represent <strong>the</strong> twoends <strong>of</strong> <strong>the</strong> spectrum <strong>of</strong> <strong>the</strong> histopathologic classification<strong>of</strong> lymphatic lesions [124]. Whe<strong>the</strong>r <strong>the</strong>se are trueneoplasms or represent malformations or hamartomas isstill debated, but this issue is <strong>of</strong> no clinical consequence.These lymphatic lesions may be divided into three morphologictypes: capillary (lymphangioma circumscriptum,ICD-O:9171/0), cavernous (lymphangioma cavernosum,ICD-O:9172/0), <strong>and</strong> cystic (cystic hygroma,ICD-O:9173/0) [34].Lymphangiomas are relatively rare. Almost alllymphangiomas appear during <strong>the</strong> first 2 years <strong>of</strong> life,most commonly in <strong>the</strong> oral cavity, parotid gl<strong>and</strong>, neckor axilla. Lymphangiomas <strong>of</strong>ten lie in <strong>the</strong> lateral cervicalregion beneath <strong>the</strong> platysma, less frequently in <strong>the</strong>anterior cervical region, <strong>and</strong> may extend into <strong>the</strong> mediastinum.Cervical cystic hygroma (hygroma colli cysticum) isa cystic lymphangioma <strong>of</strong> <strong>the</strong> neck. It can be associatedwith foetal hydrops <strong>and</strong> Turner’s syndrome [27]. Thislesion consists microscopically <strong>of</strong> lymphatic channels<strong>of</strong> variable size <strong>and</strong> shape lined with typical endo<strong>the</strong>lialcells. Focal infiltrates <strong>of</strong> lymphocytes in <strong>the</strong> stromaare common.The main differential diagnosis <strong>of</strong> lymphangiomas<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck is cavernous haemangioma.Lymphangiomas contain proteinaceous fluid <strong>and</strong> thinvalves, <strong>and</strong> <strong>the</strong> surrounding tissue is usually infiltratedby lymphocytes, whereas cavernous haemangiomas arefilled with red blood cells <strong>and</strong> lack valve structures.Treatment <strong>of</strong> lymphangioma consists <strong>of</strong> surgery;staged procedures may be required for large lesions. Recurrencerates range from 15 to 80% [38].9.4.2 HaemangiomaHaemangiomas are a heterogeneous group <strong>of</strong> vascularlesions commonly located in <strong>the</strong> head <strong>and</strong> neck. Mosthaemangiomas in this region are superficial; however,<strong>the</strong>y may arise within skeletal muscle <strong>and</strong> involve parenchymaltissue such as salivary gl<strong>and</strong>s <strong>and</strong> <strong>the</strong> thyroidgl<strong>and</strong>. Haemangiomas are classified by morphologyinto capillary, cavernous, arteriovenous, venous <strong>and</strong>epi<strong>the</strong>lioid types [78]. Of <strong>the</strong>se types, cavernous is <strong>the</strong>one that most <strong>of</strong>ten simulates a cyst. The o<strong>the</strong>r typesmore <strong>of</strong>ten manifest <strong>the</strong>mselves as solid lesions.Fig. 9.10. Mature teratoma. Cysts lined with squamous <strong>and</strong> respiratoryepi<strong>the</strong>lium. Cartilage is presentFig. 9.11. Immature teratoma composed <strong>of</strong> primitive neuroectodermaltissue9.4.3 TeratomaICD-O:9080/1Teratomas are neoplasms composed <strong>of</strong> elements frommore than one <strong>of</strong> <strong>the</strong> three germ layers (ectoderm, endoderm<strong>and</strong> mesoderm). Cervical teratomas representonly about 3% <strong>of</strong> all teratomas [9]. In <strong>the</strong> head <strong>and</strong> neckregion, lesions are also found in <strong>the</strong> central nervous system,orbit, temporal fossa, oropharynx, oral cavity, nasopharynx,nasal cavity, palate <strong>and</strong> tonsil [60].Teratomas arising in <strong>the</strong> cervical region are rare. Although<strong>the</strong>y were previously divided into those arisingfrom <strong>the</strong> thyroid gl<strong>and</strong> <strong>and</strong> those arising elsewhere, thisdistinction has not proved to be clinically useful. Themost significant clinical marker divides <strong>the</strong> tumourspresenting in infancy or early childhood from those presentingafter <strong>the</strong> first decade <strong>of</strong> life. The former groupexhibits primarily benign clinical behaviour. However,such lesions are associated with a high mortality rate at<strong>the</strong> time <strong>of</strong> birth, generally because <strong>the</strong> airway <strong>and</strong> pulmonaryfunction are compromised. The latter group is


<strong>Neck</strong> Cysts, Metastasis, Dissection Chapter 9 273composed <strong>of</strong> tumours that are usually smaller <strong>and</strong> morelikely to be malignant [9, 33, 44, 50].Various systems <strong>of</strong> classification for teratomas havebeen proposed. The majority <strong>of</strong> <strong>the</strong>se were consideredby Gonzalez-Crussi, who presented a tentative new classificationsystem for all teratomas that does not rely on<strong>the</strong> primary site <strong>of</strong> occurrence <strong>of</strong> <strong>the</strong> tumour [44].On gross examination, <strong>the</strong>se tumours are usuallycystic, but <strong>the</strong>y can be solid or multiloculated. They arecommonly encapsulated, lobulated masses that measureup to 15 cm in <strong>the</strong>ir greatest dimension [44]. On microscopicexamination, <strong>the</strong> cervical teratomas are similar tothose found in o<strong>the</strong>r anatomical regions. They may containskin, hair, fatty tissue, central nervous tissue, cartilage,bone <strong>and</strong> components <strong>of</strong> <strong>the</strong> respiratory or digestivetract (Fig. 9.10). Areas <strong>of</strong> more immature or embryonaltissue may be present (Fig. 9.11).It is exceedingly important to adequately sampleall potentially teratomatous tumours. Specifically, solidareas with necrosis or haemorrhage should be carefullyexamined. It is not unusual to find, in teratomasthroughout <strong>the</strong> body, small foci <strong>of</strong> malignant germ celltumours, especially endodermal sinus tumour or choriocarcinoma.The presence <strong>of</strong> ei<strong>the</strong>r <strong>of</strong> <strong>the</strong>se two tissuetypes adversely affects patient prognosis. It is also importantfor <strong>the</strong> pathologist to recognise that <strong>the</strong> moreimmature foetal tissues have malignant potential [10, 33,44, 50]. Patients with <strong>the</strong>se tumours require especiallyclose clinical follow-up.Cervical teratomas in <strong>the</strong> neonate are almost alwaysbenign, whereas <strong>the</strong> few reported cases <strong>of</strong> cervical teratomaarising in adults were malignant [44]. To <strong>the</strong> best<strong>of</strong> our knowledge, only seven cases <strong>of</strong> congenital cervicalteratoma with metastasis have been reported [50].Resection seems to <strong>of</strong>fer <strong>the</strong> best control in cases <strong>of</strong> aggressivebiologic behaviour.Most authors strongly favour <strong>the</strong> operative management<strong>of</strong> teratomas [33, 44, 50]. When malignant componentsare found in a teratoma, <strong>the</strong> patient may need chemo<strong>the</strong>rapy<strong>and</strong>/or radio<strong>the</strong>rapy in addition to surgery.9.4.4 Cervical SalivaryGl<strong>and</strong> Cystic NeoplasmsHeterotopic normal salivary gl<strong>and</strong>s <strong>and</strong> salivary gl<strong>and</strong>neoplasms arising in cervical lymph nodes may simulatecervical cysts [107, 131]. These are uncommon neoplasms,<strong>and</strong> <strong>the</strong> pathologist may confuse <strong>the</strong>m with metastaticsalivary gl<strong>and</strong> tumours [23, 131]. Ectopic isl<strong>and</strong>s <strong>of</strong> salivarygl<strong>and</strong> tissue within lymph nodes have been implicatedin <strong>the</strong> pathogenesis <strong>of</strong> lymphoepi<strong>the</strong>lial cysts <strong>and</strong>some neoplastic lesions by several authors [22, 108].This type <strong>of</strong> neoplasm presents as a painless mass,<strong>of</strong>ten cystic, located in <strong>the</strong> periparotid region, <strong>the</strong> upperneck, or <strong>the</strong> anterior cervical triangle. Occasionally,however, <strong>the</strong>se tumours have been described in <strong>the</strong>lower neck [108, 131]. In <strong>the</strong> series reported by Zatchuzet al., <strong>the</strong> age <strong>of</strong> <strong>the</strong> patients ranged from 10 to 81 years,with a mean <strong>of</strong> 45 years. Females were affected morecommonly than males, with a ratio <strong>of</strong> 3:1 [131].The tumours that more <strong>of</strong>ten arise in ectopic salivarygl<strong>and</strong> tissue in <strong>the</strong> lymph nodes <strong>and</strong> simulate cysts areWarthin’s tumour <strong>and</strong> sebaceous lymphadenoma. O<strong>the</strong>rrare types <strong>of</strong> salivary gl<strong>and</strong> tumours that may resemblecervical cysts, are dermal analogue tumours, mucoepidermoidcarcinomas <strong>and</strong> acinic cell carcinomas [67, 103].The pathology <strong>of</strong> <strong>the</strong>se lesions is discussed in Chap. 5.Surgical excision is <strong>the</strong> treatment <strong>of</strong> choice. In sialocarcinomas,excision <strong>of</strong> <strong>the</strong> adjacent salivary gl<strong>and</strong> mayappear to be <strong>the</strong> appropriate treatment to define <strong>the</strong> site<strong>of</strong> <strong>the</strong> primary tumour, because malignant salivary tumourslocated within lymph nodes suggest metastaticdisease.9.4.5 Miscellaneous LesionsO<strong>the</strong>r tumours that may appear as cervical cysts arecystic neurogenic neoplasms <strong>and</strong> cervical thymomas.In <strong>the</strong> neck, <strong>the</strong> most common locations for neuromaswith cystic degeneration are along <strong>the</strong> course <strong>of</strong> <strong>the</strong> vagusnerve or <strong>the</strong> cervical sympa<strong>the</strong>tic chain [4]. Cervicalthymomas are classified as being one <strong>of</strong> four types:1. Ectopic hamartomatous thymoma,2. Cervical thymoma,3. Spindle epi<strong>the</strong>lial tumour with thymus-like differentiation(SETTLE),4. Carcinoma showing thymus-like differentiation(CASTLE).Of <strong>the</strong>se, <strong>the</strong> first is benign <strong>and</strong> <strong>the</strong> second can be locallyaggressive. The third <strong>and</strong> fourth types are malignant[18].Infectious processes <strong>of</strong>ten simulate cervical cysts.Such infections can be bacterial, fungal, parasitic, or viral[46, 104, 114]. Amyloidosis <strong>and</strong> carotid artery aneurysmshave been reported to mimic cervical cystic tumours[24, 35].9.5 ParagangliomaICD-O:8680/1Paraganglioma, <strong>of</strong>ten referred to as chemodectoma, is atumour derived from paraganglia, structures <strong>of</strong> neuroectodermalcrest derivation that are found throughout<strong>the</strong> body. Paragangliomas are intimately associated withvascular <strong>and</strong> neural structures in <strong>the</strong> head <strong>and</strong> neckregion <strong>and</strong> are most commonly classified according to<strong>the</strong>ir location: jugulotympanic, vagal, carotid body, <strong>and</strong>o<strong>the</strong>rs, including laryngeal, nasal <strong>and</strong> ocular [30, 61]. In


274 M. A Luna · K. Pineda-Daboin9Fig. 9.12. Zellballen pattern in a carotid body tumourthis section, only neoplasms <strong>of</strong> <strong>the</strong> carotid body <strong>and</strong> intravagalparaganglia are discussed.Carotid body paragangliomas are <strong>the</strong> most commontumours <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck paraganglia, making up60–70% <strong>of</strong> <strong>the</strong> tumours <strong>of</strong> this type [30, 61, 100]. Lack etal. found 69 paragangliomas <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck in morethan 600,000 operations (0.12%), <strong>and</strong> only 1 in 13,400 autopsiesat Memorial Hospital in New York city [61].The tumour, typically located in <strong>the</strong> carotid bifurcation,is typically found in individuals <strong>of</strong> ei<strong>the</strong>r sex in <strong>the</strong>third to <strong>the</strong> eighth decades <strong>of</strong> life. It <strong>of</strong>ten presents as apainless, slowly enlarging mass. It is <strong>the</strong> only neoplasmthat arises in that particular location. Carotid angiographyis a valuable diagnostic aid. The risk <strong>of</strong> developingthis tumour is higher in persons living at high altitudesthan those living at sea level [100]. Carotid body paragangliomaswith endocrine activity are rare. The tumour seldomundergoes malignant transformation; histologic criteriaare <strong>of</strong> little prognostic value. The incidence <strong>of</strong> metastasisis estimated to be less than 10% [14, 61].The vagal region is <strong>the</strong> third most frequent site <strong>of</strong> involvementafter <strong>the</strong> carotid body <strong>and</strong> jugulotympanicregion. Unlike <strong>the</strong> circumscribed carotid body, <strong>the</strong> vagalparaganglioma represents collections <strong>of</strong> microscopicnests located along <strong>the</strong> vagus nerve distal to <strong>the</strong> ganglionnodosum. Because <strong>of</strong> <strong>the</strong> variability in location <strong>of</strong> <strong>the</strong>normal vagal paraganglia, paragangliomas arising from<strong>the</strong>se structures also vary in location. At <strong>the</strong> time <strong>of</strong> diagnosis,patients are usually in <strong>the</strong> fourth to fifth decade<strong>of</strong> life, <strong>and</strong> <strong>the</strong>re is a female predominance [14, 30].Patients generally report a slowly growing neck mass,<strong>and</strong>, because <strong>of</strong> <strong>the</strong> intimate relationship with <strong>the</strong> vagusnerve, cranial nerve palsies may also be present [14, 30].Vagal paragangliomas displace <strong>the</strong> carotid vessels anteriorly,are grossly round or fusiform, <strong>and</strong> abut <strong>the</strong> base<strong>of</strong> <strong>the</strong> skull. Multiple, also bilateral, <strong>and</strong> familial occurrences<strong>of</strong> paragangliomas have been documented.Paragangliomas have a tan, s<strong>of</strong>t cut surface. Paragangliomasfrom all regions <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck arehistologically similar. They are well circumscribed <strong>and</strong>composed <strong>of</strong> chief cells arranged in nests known as Zellballen(Fig. 9.12). The tumour cells have granular cytoplasm<strong>and</strong> round nuclei with prominent nucleoli. Nuclearpleomorphism may be present, but mitosis is rare, <strong>and</strong>necrosis is usually present only if <strong>the</strong> patient underwentpreoperative embolisation or if <strong>the</strong> nests <strong>of</strong> cells are verylarge. Compressed sustentacular cells <strong>and</strong> a rich capillarynetwork surround each nest. A reticulin stain highlights<strong>the</strong> Zellballen arrangement <strong>of</strong> <strong>the</strong> cells. Malignantvarieties are difficult to distinguish on histologic examination,but generally <strong>the</strong>y have a higher mitotic rate <strong>and</strong>more necrosis than benign tumours. Vascular invasionmay be present in both benign <strong>and</strong> malignant paragangliomas.The chief cells are positive for neuroendocrine markerssuch as chromogranin <strong>and</strong> synaptophysin. They areusually negative for cytokeratin, but an occasional casehas been reported to be positive [55]. The sustentacularcells are positive for S-100 protein [55].Electron microscopy studies show <strong>the</strong> tumour cellsto contain neurosecretory granules. The cells have cytoplasmicprocesses that surround neighbouring cells,<strong>and</strong> <strong>the</strong> cytoplasm contains abundant large mitochondria<strong>and</strong> inconspicuous Golgi apparatus, smooth <strong>and</strong>rough endoplasmic reticulum.Although histologic findings are generally quite distinctive,<strong>the</strong> differential diagnosis <strong>of</strong> paragangliomas <strong>of</strong><strong>the</strong> head <strong>and</strong> neck may include endocrine neoplasms arisingfrom <strong>the</strong> thyroid (medullary carcinoma) or parathyroidgl<strong>and</strong>s <strong>and</strong> o<strong>the</strong>r neuroendocrine carcinomas. Lesscommonly, alveolar s<strong>of</strong>t part sarcoma, melanoma, granularcell tumour <strong>and</strong> metastatic renal cell carcinoma are includedin <strong>the</strong> differential diagnosis [14, 30, 55, 61].Surgery is <strong>the</strong> treatment <strong>of</strong> choice for paragangliomas.If <strong>the</strong> neoplasm is completely excised, recurrence isrelatively rare; recurrence rates are 10% for carotid bodytumours <strong>and</strong> 5–25% for vagal paragangliomas [14, 30,55, 61]. Radio<strong>the</strong>rapy may be useful as a palliative methodfor those tumours that cannot be controlled by surgicalmeans. Local infiltration <strong>of</strong> vagal body tumours <strong>and</strong>extension into <strong>the</strong> cranial cavity represent significantproblems in disease control. The rate <strong>of</strong> metastasis in intravagaltumours is estimated at 16%, but most <strong>of</strong> <strong>the</strong>seare to regional lymph nodes [14, 30].9.6 Unknown Primary<strong>and</strong> Secondary Tumours9.6.1 DefinitionIn head <strong>and</strong> neck oncology, <strong>the</strong> term “ unknown primarytumour” means a primary neoplasm that has not beenfound in a patient with neck metastasis, even after a


<strong>Neck</strong> Cysts, Metastasis, Dissection Chapter 9 275Table 9.2. Location <strong>of</strong> lymph node metastasis <strong>and</strong> predominant sites <strong>of</strong> <strong>the</strong>ir primary tumours (extracted from [75, 99, 125]. ENT earnose throat, GI gastrointestinal, GU genitourinaryLymph node region affected by metastasisSublevel IA (submental)Sublevel IB (subm<strong>and</strong>ibular)Sublevel IIA (upper jugular)Sublevel IIB (upper jugular)Level III (middle jugular)Level IV (lower jugular)Sublevels VA, VB (posterior cervical)SupraclavicularPredominant sites(s) <strong>of</strong> primary tumourAnterior floor <strong>of</strong> mouth; anterior oraltongue, anterior m<strong>and</strong>ibular ridge lower lipOral cavity, anterior nasal cavity,midface, subm<strong>and</strong>ibular gl<strong>and</strong>Waldeyer’s ring, oral cavity, nasalcavity, oropharynx, supraglottis, floor<strong>of</strong> mouth, pyriform sinusAnterior tongue, nasopharynx, tonsilHypopharynx, base <strong>of</strong> tongueposterior pharyngeal wall, supraglottic larynxHypopharynx, thyroidNasopharynx, thyroid, oropharynxLungs (40%), thyroid (22%)GI tract (12%), GU tract (8%)all ENT regions (20%)thorough work-up. The neck metastasis may representregional or distant primary disease.shows <strong>the</strong> probable primary sites by region <strong>of</strong> metastasis[75, 109, 125].9.6.2 Clinical FeaturesAn enlarged cervical lymph node is frequently <strong>the</strong> firstclinical manifestation <strong>of</strong> a neoplastic process in <strong>the</strong> head<strong>and</strong> neck. Cervical lymph node metastasis is <strong>the</strong> presentingsymptom in 25% <strong>of</strong> patients with cancer <strong>of</strong> <strong>the</strong>oral cavity or pharynx, in 47% <strong>of</strong> patients with nasopharyngealcarcinoma, <strong>and</strong> in 23% <strong>of</strong> patients with thyroidcarcinoma. In some instances, however, despite a thoroughsearch, a primary tumour cannot be found [45, 64,72, 79, 93, 123].Patients with a high probability <strong>of</strong> a metastatic tumourin <strong>the</strong> cervical lymph nodes are men (male: femaleratio 4:1) older than 40 years, who smoke <strong>and</strong>drink alcohol heavily. They usually present with apainless node larger than 2 cm along <strong>the</strong> jugular chainor <strong>the</strong> supraclavicular fossa. Various groups <strong>of</strong> lymphnodes can be affected by metastatic neoplasms, but <strong>the</strong>most frequently involved are <strong>the</strong> upper jugular (71%),<strong>the</strong> midjugular (22%), <strong>the</strong> supraclavicular (18%), <strong>and</strong><strong>the</strong> posterior cervical nodes (12%). Approximately 14%<strong>of</strong> patients with such disease have more than one lymphnode group affected by metastases <strong>and</strong> 10% have bilaterallymph node metastases [45, 123]. The lymphaticdrainage <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck region is highly predictable,<strong>and</strong> <strong>the</strong> location <strong>of</strong> <strong>the</strong> adenopathy may providea clue to <strong>the</strong> location <strong>of</strong> <strong>the</strong> primary lesion. Table 9.29.6.3 Searchfor <strong>the</strong> Primary TumourIf <strong>the</strong> search is conducted systematically, <strong>the</strong> primarycancer can be discovered in 75–90% <strong>of</strong> patients presentingwith cervical adenopathy. Evaluation protocols havebeen outlined by several authors [66, 72]. With advancesin upper aerodigestive tract examination that use flexiblefibre optic endoscopes <strong>and</strong> rigid telescopes, <strong>the</strong> failure<strong>of</strong> radiographic tests to determine <strong>the</strong> location <strong>of</strong> anunknown primary has become even more apparent. Thismay change in <strong>the</strong> future, however, with <strong>the</strong> application<strong>of</strong> advanced radiologic techniques that allow functionalassessment <strong>of</strong> tissues. Single-photon emission computedtomography (SPECT) using 2-(18-F) fluoro-2-deoxy-D-glucose was able to detect 9 out <strong>of</strong> 11 histologicallyproven occult primary neoplasms <strong>of</strong> <strong>the</strong> upper aerodigestivetract [77].The success rates <strong>of</strong> <strong>the</strong> ultimate detection <strong>of</strong> occultprimaries (by various authors) vary between 10<strong>and</strong> 75% [64, 72, 79]. As might be expected, occultprimary tumours are significantly less likely to be detectedin patients treated with radiation [98]. Nearly50% <strong>of</strong> primary carcinomas originally considered occultthat are eventually found in patients after treatment<strong>of</strong> <strong>the</strong> cervical lymph node are located in <strong>the</strong> region<strong>of</strong> Waldeyer’s ring [73, 116]. Of originally un-


276 M. A Luna · K. Pineda-DaboinTable 9.3. Frequency <strong>of</strong> histologic type <strong>of</strong> metastases from unknown primary tumours (Extracted from [64, 66, 75])CervicalSquamous cell carcinomaUndifferentiated carcinomaMelanomaThyroid carcinomaAdenocarcinomaSalivary gl<strong>and</strong> carcinomaSupraclavicularAdenocarcinomaSquamous cell carcinomaUndifferentiated carcinomaThyroid carcinomaProstate carcinomaSarcoma9known primary tumours found below <strong>the</strong> clavicle, <strong>the</strong>largest number are in <strong>the</strong> lungs, followed by <strong>the</strong> gastrointestinaltract.In cases in which <strong>the</strong> primary tumour is not found,<strong>the</strong> most likely explanations are that ei<strong>the</strong>r <strong>the</strong> primarylesion is so small that it is not visible or it has regressedspontaneously. The latter consideration is speculative,since spontaneous regression cannot be proved. Although<strong>the</strong> origin <strong>of</strong> a carcinoma in <strong>the</strong> wall <strong>of</strong> a branchiogeniccyst is possible, this remains entirely hypo<strong>the</strong>tical.Few observers are willing to agree that primarysquamous cell carcinoma (SCC) or o<strong>the</strong>r types <strong>of</strong> carcinomaarise in branchiogenic cysts. The most attractivehypo<strong>the</strong>sis, <strong>the</strong>refore, is that <strong>the</strong> primary neoplasm istoo small to be detected.9.6.4 Common Location<strong>of</strong> <strong>the</strong> Primary TumourThe gross appearance <strong>of</strong> a metastasis is important onlywhen it is present in a cystic node, because in cystic metastases,<strong>the</strong> primary tumour is most frequently locatedin <strong>the</strong> palatine tonsils or in Waldeyer’s ring [79, 96, 116].In o<strong>the</strong>r situations, <strong>the</strong> metastatic deposits may adopt amultitude <strong>of</strong> appearances, from <strong>the</strong> solid white-grey toyellow haemorrhagic deposits <strong>of</strong> renal cell carcinoma to<strong>the</strong> deeply pigmented focus <strong>of</strong> metastatic melanoma.9.6.5 Histologic Type <strong>of</strong> Metastases<strong>and</strong> ImmunohistochemicalFeaturesNearly any histologic type <strong>of</strong> malignancy can present asa metastasis to <strong>the</strong> cervical lymph nodes, but metastaticSCC is by far <strong>the</strong> most common tumour to do so. In cases<strong>of</strong> metastases <strong>of</strong> an unknown primary tumour to <strong>the</strong>cervical lymph nodes, 80–85% are <strong>of</strong> this histologic type(Table 9.3). Undifferentiated carcinomas, adenocarcinomas,thyroid carcinomas, melanoma, rhabdomyosarcomas<strong>and</strong> sialocarcinomas are <strong>the</strong> o<strong>the</strong>r neoplasms that<strong>of</strong>ten metastasise to cervical lymph nodes. Adenocarcinomas,undifferentiated carcinomas <strong>and</strong> thyroid carcinomasmore commonly metastasise to <strong>the</strong> supraclavicular<strong>and</strong> scalene nodes. Most <strong>of</strong> <strong>the</strong> adenocarcinomas in<strong>the</strong> upper jugular region are metastases from lesions <strong>of</strong><strong>the</strong> sinonasal tract or salivary gl<strong>and</strong>s.The origin <strong>of</strong> a keratinising SCC, regardless <strong>of</strong> differentiation,cannot be suggested by its morphology alone.The location <strong>of</strong> <strong>the</strong> node can, however, be a clue about <strong>the</strong>location <strong>of</strong> <strong>the</strong> primary neoplasm (Table 9.2). The cytokeratin(CK) pattern may be <strong>of</strong> some help in determining<strong>the</strong> origin <strong>of</strong> <strong>the</strong> metastasis. SCC <strong>of</strong> <strong>the</strong> upper aerodigestivetract are positive for CK 5/6, 10, 13,14 ,17 <strong>and</strong> 19,whereas SCC <strong>of</strong> <strong>the</strong> lung are positive for CK 5/6, 12 <strong>and</strong>14 in 100% <strong>of</strong> cases <strong>and</strong> CK 17, 8/18 <strong>and</strong> 19 in 80% <strong>of</strong> cases.Fewer than 4% <strong>of</strong> cases are positive for CK 7 <strong>and</strong> 20[21]. Fur<strong>the</strong>rmore, thyroid transcription factor 1 (TTF-1)is positive in 10 to 37% <strong>of</strong> pulmonary SCC [83].A cystic neoplasm composed <strong>of</strong> poorly differentiatednon-keratinising carcinoma recapitulating tonsillarcrypt epi<strong>the</strong>lium (Fig. 9.13) most likely originates in <strong>the</strong>lingual or faucial tonsil [73, 96, 116]. Metastases from<strong>the</strong> tonsil are <strong>of</strong>ten unicystic, whereas those from <strong>the</strong>tongue are more <strong>of</strong>ten multicystic [96]. Since <strong>the</strong> carcinomasare deep in <strong>the</strong> tonsils, tonsillectomy ra<strong>the</strong>rthan biopsy is needed to demonstrate <strong>the</strong> primary neoplasm[97]. A subset <strong>of</strong> <strong>the</strong>se crypt carcinomas are <strong>of</strong>tenpositive for CK 7, especially those with basaloid features(Fig. 9.14) [94]. These two types <strong>of</strong> metastatic cysticcarcinoma are <strong>of</strong>ten mistaken for a branchial cleftcyst or branchogenic carcinomas by <strong>the</strong> unwary pathologist[73, 96, 116].9.6.6 Differential DiagnosisCystic metastatic SCC should be distinguished frombenign lesions lined with benign squamous epi<strong>the</strong>lium,such as branchial cleft cysts, AIDS-related cystic lymphoidhyperplasia, benign lymphoepi<strong>the</strong>lial cysts, thymiccysts <strong>and</strong> cystic cervical thymomas. In all <strong>the</strong>se lesions,<strong>the</strong> bl<strong>and</strong> appearance <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium rules outmetastatic SCC.The most common location <strong>of</strong> metastatic adenocarcinomasin <strong>the</strong> neck is in <strong>the</strong> lower regions, <strong>and</strong> <strong>the</strong> primaryneoplasms are usually located in <strong>the</strong> thyroid, lung,


<strong>Neck</strong> Cysts, Metastasis, Dissection Chapter 9 277Fig. 9.13. Metastatic cystic tonsillar crypt carcinoma, resemblinga branchial cleft cyst. Inset: high power view <strong>of</strong> non-keratinisingcarcinoma, human papilloma virus type 16 positiveFig. 9.14. Cytokeratin 7 positive metastatic tonsillar basaloid carcinomain a cervical lymph nodeTable 9.4. Immunohistochemical approach to metastasis <strong>of</strong> unknown primary tumour. (Extracted from references [19, 21, 29 ,40, 56,66, 83, 84, 94])Cytokeratin-positiveKeratinizing squamous cell carcinoma CKs 5/6, 7 <strong>and</strong> 20Non-keratinizing squamous carcinoma CKs 5/6, 7 <strong>and</strong> 20EBV, HPV 16Adenocarcinoma CKs 7, 20TTF-1ThyroglobulinFemales: GCDFP-15, WT-1, CA 125Males: PSA .Young males: AFP, HCGUndifferentiated carcinoma CKs 7 <strong>and</strong> 20SynaptophysinChromograninEBVCytokeratin-negativeVarious tumoursS-100 proteinHMB 45Melan ADesminLymphoma markersgastrointestinal tract, or prostate. In <strong>the</strong> upper <strong>and</strong> middleneck, on <strong>the</strong> o<strong>the</strong>r h<strong>and</strong>, <strong>the</strong> primary lesions are locatedin <strong>the</strong> sinonasal tract <strong>and</strong> salivary gl<strong>and</strong>s. Only incases <strong>of</strong> thyroid carcinoma or prostate carcinoma is <strong>the</strong>origin <strong>of</strong> an adenocarcinoma apparent from <strong>the</strong> morphology<strong>of</strong> <strong>the</strong> nodal metastases. Metastatic adenocarcinomaswith enteric morphology can arise in <strong>the</strong> sinonasalregion; <strong>the</strong>y are CK 20-positive, like <strong>the</strong>ir counterparts<strong>of</strong> colonic origin [21]. Thyroglobulin, calcitonin,<strong>and</strong> TTF-1 are useful markers to probe <strong>the</strong> thyroid origin<strong>of</strong> a neoplasm <strong>of</strong> unknown origin [83]. Adenocarcinoma<strong>of</strong> <strong>the</strong> prostate may present as metastasis in <strong>the</strong> leftside <strong>of</strong> <strong>the</strong> neck, especially in <strong>the</strong> supraclavicular nodes.The diagnosis can be confirmed by using <strong>the</strong> prostatespecificantigen (PSA) test (Table 9.4).The presence <strong>of</strong> oestrogen receptors <strong>and</strong> gross cysticdisease fluid protein 15 (GCDFP-15) would suggest abreast origin for adenocarcinoma, but <strong>the</strong>se markers arenon-specific for <strong>the</strong> breast. Lung adenocarcinomas arepositive for TTF-1 <strong>and</strong> B 72.3 (Table. 9.4) [83].Benign gl<strong>and</strong>ular inclusions in cervical lymph nodesshould not be mistaken for metastatic adenocarcinomas;heterotopic gl<strong>and</strong>s <strong>of</strong> salivary tissue are commonin <strong>the</strong> paraparotid lymph nodes <strong>and</strong> less common in <strong>the</strong>upper cervical nodes. Acinic cell carcinomas, mucoepidermoidcarcinomas, Warthin’s tumour <strong>and</strong> pleomorphicadenomas have been described in cervical lymphnodes, <strong>and</strong> <strong>the</strong>y should not be confused with metastaticadenocarcinomas [23, 67, 131].


278 M. A Luna · K. Pineda-Daboin9Metastatic spindle cell neoplasms most likely representsarcomatoid carcinomas, melanomas <strong>and</strong> sarcomas,especially rhabdomyosarcomas. Pankeratin, MART-1, HMB-45, S-100 protein, desmin, smooth muscle actin<strong>and</strong> myogenin are some immunostains that help todistinguish <strong>the</strong>se neoplasms [29]. Primary spindle celllesions arising in lymph nodes, such as Kaposi’s sarcoma,presumed tumours <strong>of</strong> <strong>the</strong> reticulum cell lineage <strong>and</strong>benign intranodal my<strong>of</strong>ibroblastomas, must be distinguishedfrom metastatic spindle neoplasms [2, 56].Undifferentiated malignant neoplasms in cervicallymph nodes need to be investigated with immunohistochemicalstudies. CK-positive malignancies with <strong>the</strong>morphology <strong>of</strong> <strong>the</strong> nasopharyngeal type <strong>of</strong> carcinomas(NPC) are usually located in <strong>the</strong> posterior neck, whereasmetastasis from undifferentiated sinonasal carcinomas(SNUC) are present in <strong>the</strong> upper <strong>and</strong> mid-cervical regions.If <strong>the</strong> metastases are located in <strong>the</strong> lower neck, <strong>the</strong>lung is <strong>the</strong> most likely source. If <strong>the</strong> carcinomas in additionexhibited neuroendocrine differentiation, Merkelcell carcinoma or small cell neuroendocrine carcinoma,from ei<strong>the</strong>r <strong>the</strong> lung or <strong>the</strong> larynx, is <strong>the</strong> best diagnosis.Merkel cell carcinomas are CK 20-positive [19] <strong>and</strong> pulmonarysmall cell carcinomas are CK 20-negative <strong>and</strong>TTF-1 positive in 83–100% <strong>of</strong> cases [83]. Benign nevuscells have been found in <strong>the</strong> capsules <strong>of</strong> subm<strong>and</strong>ibularlymph nodes. This rare finding should not lead to an erroneousdiagnosis <strong>of</strong> malignancy [54].Paccioni et al. evaluated 25 cases <strong>of</strong> occult metastasisto cervical lymph nodes for <strong>the</strong> presence <strong>of</strong> Epstein-Barr virus (EBV) by in situ hybridisation following fineneedleaspiration biopsies <strong>of</strong> <strong>the</strong> neck mass <strong>and</strong> correlated<strong>the</strong> findings with <strong>the</strong> histologic types <strong>of</strong> <strong>the</strong> surgicalspecimens (after locating <strong>the</strong> primary site <strong>of</strong> origin).These authors reported that EBV was expressed in 7 metastases,ultimately proving <strong>the</strong>ir origin from <strong>the</strong> nasopharynx,while <strong>the</strong> remaining 18 cases (not <strong>of</strong> Waldeyer’sring origin) were negative for EBV [84]. The authorsindicated that detection <strong>of</strong> EBV in cervical metastasesmay assist in <strong>the</strong> localisation <strong>of</strong> <strong>the</strong> occult primary toWaldeyer’s ring. SNUC do not express EBV [84].The pattern <strong>of</strong> CK expression is significantly differentin SNUC <strong>and</strong> NPC, which could be <strong>of</strong> diagnosticaid. Franchi et al. demonstrated that SNUC expressCK 8 in 100% <strong>of</strong> cases <strong>and</strong> CK 19 <strong>and</strong> CK 7 in 50%, <strong>and</strong>are negative for CK 5/6, while NPC express CK 5/6 <strong>and</strong>CK 13 in 90% <strong>of</strong> cases <strong>and</strong> are negative for CK 7 [40].Metastases from melanoma, rhabdomyosarcoma,<strong>and</strong> rarely from olfactory neuroblastoma should beconsidered with <strong>the</strong> CK-negative undifferentiated neoplasms<strong>and</strong> proper immunohistochemical markers investigated(Fig. 9.15). In CK-negative tumours, <strong>the</strong> possibility<strong>of</strong> malignant lymphoma should be considered<strong>and</strong> CD 20, CD 3 <strong>and</strong> antibodies for leukocyte commonantigen should be measured [29].Fig. 9.15. Metastatic spindle cell melanoma in a cervical lymphnode9.6.7 Treatment <strong>and</strong> ResultsFactors that affect survival in patients with metastasisfrom an unknown primary tumour are clinical stage<strong>of</strong> <strong>the</strong> neck, extranodal extension <strong>of</strong> metastasis <strong>and</strong> <strong>the</strong>presence <strong>of</strong> recurrent or residual disease after treatment.Surgery combined with irradiation has resultedin better local control <strong>of</strong> <strong>the</strong> disease than ei<strong>the</strong>r <strong>the</strong>rapyalone [53]. The 5-year survival results in series <strong>of</strong>patients with SCC metastatic to <strong>the</strong> neck from an unknownprimary origin range from 0 to 54% [45, 64, 72,75]. In <strong>the</strong> last series from <strong>the</strong> University <strong>of</strong> Texas M.D.Anderson Cancer Center, <strong>the</strong> 5-year actuarial survivalrate was 55% [123].9.7 <strong>Neck</strong> Dissection9.7.1 Classification<strong>of</strong> <strong>Neck</strong> DissectionsThe American Academy <strong>of</strong> Otolaryngology-<strong>Head</strong> <strong>and</strong><strong>Neck</strong> Surgery <strong>and</strong> <strong>the</strong> American Society <strong>of</strong> <strong>Head</strong> <strong>and</strong><strong>Neck</strong> Surgery classified neck dissection into four categories:radical, modified radical, extended <strong>and</strong> selective[99].<strong>Neck</strong> dissection is classified primarily by <strong>the</strong> cervicallymph node groups that are removed, <strong>and</strong> secondarilyon <strong>the</strong> anatomic structures that may be preserved, suchas <strong>the</strong> spinal accessory nerve, <strong>the</strong> sternocleidomastoidmuscle, <strong>and</strong> <strong>the</strong> internal jugular vein [71, 99].The cervical lymph node groups are referred to using<strong>the</strong> level system as described by <strong>the</strong> Sloan-Kettering MemorialHospital Group (Fig. 9.1). Definition <strong>of</strong> <strong>the</strong> anatomicboundaries <strong>of</strong> <strong>the</strong> different lymph node groupsis beyond <strong>the</strong> scope <strong>of</strong> this chapter <strong>and</strong> can be found ino<strong>the</strong>r sources [99, 110].


<strong>Neck</strong> Cysts, Metastasis, Dissection Chapter 9 279Table 9.5. Updated classification <strong>of</strong> neck dissection (extracted from [99]). SCM sternocleidomastoid, IJV internal jugular vein, SANspinal accessory nerveType <strong>of</strong> dissection Lymph node levels removed Non-lymphatic structures resectedRadical neck dissection I, II, III, IV, V SCM, IJV, SANModified radical neck dissection I, II, III, IV, V Preservation <strong>of</strong> one or more <strong>of</strong> <strong>the</strong> following:SCM, IJV, SANSelective neck dissection Preservation <strong>of</strong> one or Nonemore <strong>of</strong> <strong>the</strong> following:I, II, III, IV, V. Bracketsare used to denote levelsor sublevels removed. (e.g.SND {I, II, III})Extended neck dissection Resection <strong>of</strong> one or more Resection <strong>of</strong> one or moreor additional lymph nodes non-lymphatic structureslevels routinely notroutinely not removed byremoved by <strong>the</strong> radical<strong>the</strong> radical neck dissectionneck dissection(e.g. carotid artery)(e.g. parapharyngeal)Radical neck dissection consists <strong>of</strong> <strong>the</strong> removal <strong>of</strong> allfive lymph node regions <strong>of</strong> one side <strong>of</strong> <strong>the</strong> neck ( levelsI–V). This includes removal <strong>of</strong> <strong>the</strong> sternocleidomastoidmuscle, <strong>the</strong> internal jugular vein, <strong>and</strong> <strong>the</strong> spinal accessorynerve. Modified radical neck dissection refers to excision<strong>of</strong> all lymph nodes routinely removed by radicalneck dissection, with preservation <strong>of</strong> one or more <strong>of</strong> <strong>the</strong>non-lymphatic structures (i.e. spinal accessory nerve,internal jugular vein, <strong>and</strong>/or sternocleidomastoid muscle).The term extended radical neck dissection refers to aneck dissection that is extended to include ei<strong>the</strong>r lymphnode groups or non-lymphatic structures that are notroutinely removed in a st<strong>and</strong>ard radical neck dissection.Selective neck dissection is any type <strong>of</strong> cervicallymphadenectomy in which one or more <strong>of</strong> <strong>the</strong> lymphnode groups that are removed in a radical neck dissectionis preserved [13, 71, 99] (Table 9.5).9.7.2 Gross Examination<strong>of</strong> <strong>Neck</strong> DissectionSurgical SpecimensThe following procedure pertains to st<strong>and</strong>ard radicalneck dissections <strong>and</strong> needs to be modified for <strong>the</strong> o<strong>the</strong>rthree types. When <strong>the</strong> main anatomic l<strong>and</strong>marks suchas <strong>the</strong> subm<strong>and</strong>ibular gl<strong>and</strong> <strong>and</strong> internal jugular veinare lacking in a neck dissection specimen, <strong>the</strong> surgeonmust identify <strong>and</strong> label <strong>the</strong> lymph node groups. This isespecially important in selective <strong>and</strong> extended neck dissections.After <strong>the</strong> neck dissection specimen has been orientedas it appears in vivo, its overall dimensions are measured.The lengths <strong>of</strong> <strong>the</strong> sternocleidomastoid muscle <strong>and</strong> <strong>the</strong>internal jugular vein are measured separately. The jugularvein should be opened along its entire length. Tumourinvolvement, including thrombosis, should benoted, described <strong>and</strong> sampled adequately. Next, <strong>the</strong>subm<strong>and</strong>ibular gl<strong>and</strong>, <strong>the</strong> sternocleidomastoid muscle,<strong>and</strong> <strong>the</strong> internal jugular vein should be divided, <strong>and</strong> <strong>the</strong>node-containing fat separated into <strong>the</strong> five levels:1. Sublingual <strong>and</strong> subm<strong>and</strong>ibular,2. Superior jugular,3. Middle jugular,4. Inferior jugular,5. Posterior.The presence <strong>of</strong> tumour in s<strong>of</strong>t tissues, subm<strong>and</strong>ibulargl<strong>and</strong> <strong>and</strong> muscle should be described. All lymph nodesvisible <strong>and</strong> palpable are carefully dissected from connectivetissue with a rim <strong>of</strong> perinodal connective tissueor fat. The number <strong>of</strong> lymph nodes (by level) should benoted; if a tumour is present, <strong>the</strong> actual size <strong>of</strong> metastasesin centimetres <strong>and</strong> <strong>the</strong> presence <strong>of</strong> extracapsularextension are also noted <strong>and</strong> recorded. It is generallyrecognised that most masses larger than 3 cm in diameterare not single nodes but are confluent nodes ortumour in s<strong>of</strong>t tissues [7]. Midline nodes, if found, areconsidered ipsilateral nodes. The contralateral neck dissectionis treated similarly. Nodes larger than 2–3 cm


280 M. A Luna · K. Pineda-Daboin9are bisected along <strong>the</strong>ir longest axis plane <strong>and</strong> bothhalves submitted. Smaller lymph nodes are submittedin toto. The tissue sections <strong>of</strong> lymph nodes submittedfor processing should include a capsule <strong>of</strong> lymph node,including a rim <strong>of</strong> perinodal connective tissue or fat. Ifa group <strong>of</strong> matted lymph nodes is present, two or threesections through <strong>the</strong> nodes are <strong>of</strong>ten adequate to document<strong>the</strong> extent <strong>of</strong> <strong>the</strong> tumour. Tissue sections submittedfor processing include all lymph nodes (by level), <strong>the</strong>subm<strong>and</strong>ibular gl<strong>and</strong>, <strong>the</strong> sternocleidomastoid muscle,<strong>and</strong> <strong>the</strong> internal jugular vein. If <strong>the</strong> neck dissection is<strong>of</strong> <strong>the</strong> extended type, sections <strong>of</strong> all extra lymph nodegroups <strong>and</strong> non-lymphatic structures that were removedshould be submitted for tissue processing.9.7.3 Histologic Evaluation<strong>of</strong> <strong>Neck</strong> DissectionThe major aim <strong>of</strong> <strong>the</strong> histologic evaluation <strong>of</strong> <strong>the</strong> status<strong>of</strong> lymph nodes in cases <strong>of</strong> carcinoma <strong>of</strong> <strong>the</strong> head<strong>and</strong> neck is to provide information required for stagingdisease, planning fur<strong>the</strong>r treatment <strong>and</strong> predictingpatient outcome. The histologic evaluation alsodocuments <strong>and</strong> confirms <strong>the</strong> pathologist’s own grossevaluation <strong>of</strong> <strong>the</strong> dissected specimen. More importantly,increasingly important histologic parameters,such as number, sizes <strong>and</strong> levels <strong>of</strong> positive nodes, <strong>the</strong>presence or absence <strong>of</strong> extracapsular spread, <strong>the</strong> presence<strong>of</strong> desmoplastic reaction, <strong>the</strong> presence <strong>of</strong> gross residualtumour <strong>and</strong> <strong>the</strong> presence <strong>of</strong> tumour emboli inintervening lymphatics, among o<strong>the</strong>rs, can be assessedby a thorough histologic evaluation. These histologicfindings by <strong>the</strong>mselves <strong>and</strong> in combination with o<strong>the</strong>rhistologic parameters have been increasingly identifiedas important prognostic factors in disease control<strong>and</strong> survival, recurrence <strong>of</strong> neck disease <strong>and</strong> distantmetastasis. The five factors that are currently indicatorsfor adjuvant postoperative <strong>the</strong>rapy can be reliablyprovided only by histologic evaluation; <strong>the</strong>y are:1. Extranodal spread <strong>of</strong> disease,2. Number <strong>of</strong> lymph nodes involved,3. Number <strong>of</strong> lymph node regions involved,4. Size <strong>of</strong> metastases,5. Presence <strong>of</strong> desmoplastic reaction in metastatic disease[42].References1. Agaton-Bonilla FC, Gay-Escola C (1996) Diagnosis <strong>and</strong>treatment <strong>of</strong> branchial cleft cysts <strong>and</strong> fistulae. A retrospectivestudy <strong>of</strong> 183 patients. Oral Maxill<strong>of</strong>ac Surg 25:449–4452. Aguacil-Garcia A (1992) Intranodal my<strong>of</strong>ibroblastoma in asubm<strong>and</strong>ibular lymph node. A case report. Am J Clin Pathol978:69–723. Al-Dousary S (1997) Current management <strong>of</strong> thyroglossalductremnant. J Otolaryngol 126:259–2654. Al-Gh<strong>and</strong>i S, Black MJ, Lafond G (1992) Extracranial head<strong>and</strong> neck schwannomas. J Otolaryngol 2:86–885. Allard RHB (1982) The thyroglossal duct cyst. <strong>Head</strong> <strong>Neck</strong>1:134–1366. Aluffi P, Pino M, Boldorin R et al. (2003). Papillary thyroidcarcinoma identified after Sistrunk procedure, report <strong>of</strong> twocases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Tumori 89:207–2107. American Joint Committee on Cancer (1997) Cancer stagingmanual. Lippincott, Philadelphia8. Ayache D, Ducroz V, Roger G et al. (1997) Midline cervicalcleft. Int Pediatr Otorhinolaryngol 40:189–1939. Batsakis JG, El-Naggar AK, Luna MA (1995) Teratomas <strong>of</strong><strong>the</strong> head <strong>and</strong> neck with emphasis on malignancy. Ann OtolRhinol Laryngol 104:456–50010. Batsakis JG, El Naggar AK, Luna MA (1996) Thyroid gl<strong>and</strong>ectopia. Ann Otol Rhinol Laryngol 105:996–100011. Baurmash HD (2003) Mucoceles <strong>and</strong> ranulas. J Oral Maxill<strong>of</strong>acSurg 61:369–37812. Bielamowicz SA, Storper BA et al. (1994) Spaces <strong>and</strong> triangles<strong>of</strong> head <strong>and</strong> neck. <strong>Head</strong> <strong>Neck</strong> 164:383–38813. Byers RM (1985) Modified neck dissection: a study <strong>of</strong> 967cases from 1970 to 1980. Am J Surg 150:414–42114. Carlsen CS, Godballe C, Krogdahl AS et al. (2003). Malignantvagal paraganglioma: report <strong>of</strong> a case treated with embolization.Auris Nasus Larynx 30:443–44615. Carroll W, Zappia JL, McClatchey K (1993) Branchiogeniccarcinoma. J Otolaryngol 22:26–2816. Cassano L, Lombardo P, Marchese-Ragona R et al. (2000).Laryngopyocele: three new cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.Eur Arch Otorhinolaryngol 257:507–51117. Cassarino DS, Milas M, Folpe AL (2003) Bilateral intrathyroidallymphoepi<strong>the</strong>lial cyst. Arch Pathol Lab Med 127:251–25218. Chan JK, Rosai J (1991) Tumors <strong>of</strong> <strong>the</strong> neck showing thymicor related brachial pouch differentiation. A unifying concept.Hum Pathol 22:349–36719. Chan JKC, Suster S, Wenig B et al. (1997) Cytokeratin 20 immunoreactivitydistinguishes Merkel cell (primary cutaneousneuroendocrine) carcinomas <strong>and</strong> salivary gl<strong>and</strong> smallcell carcinomas from small cell carcinomas <strong>of</strong> various sites.Am J Surg Pathol 21:226–23420. Choi SS, Zalzal GH (1995). Branchial anomalies a review <strong>of</strong>52 cases. Laryngoscope 105:909–91321. Chu PG, Weiss LM (2002) Keratin expression in human tissue<strong>and</strong> neoplasms. Histopathology 40:403–43922. Cleary K, Batsakis JG (1990) Lymphoepi<strong>the</strong>lial cyst <strong>of</strong> <strong>the</strong> parotidgl<strong>and</strong> region. A new face on an old lesion. Ann OtolRhinol Laryngol 99:62–6423. Croce A, Moreti H, Bianchedi M et al. (1996) Carcinoma inectopic Warthin’s tumor: a case study <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature.Acta Otorhinolaryngol Ital 16:543–54924. Cunningham MJ, Rueger RG, Rothfus WE (1989) Extracranialcarotid aneurysm: an unusual neck mass in a youngadult. Ann Otol Rhinol Laryngol 98:396–39925. Damiano A, Glickman AB, Rubin JS et al. (1996) Ectopic thyroidtissue presenting as a midline neck mass. Int J PediatrOtorhinolaryngol 34:141–14826. Davenport M (1996) Lumps <strong>and</strong> swelling <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck. BMJ 312:368–37127. De Serres L, Sie KCY, Richardson MA (1995) Lymphaticmalformations <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. A proposal for staging.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 121:577–58228. Deane SA, Tel<strong>and</strong>er RL (1978) Surgery for thyroglossal duct<strong>and</strong> branchial cleft anomalies. Am J Surg 136:348–35329. DeYoung BR, Wick MR (2000) Immunohistochemical evaluation<strong>of</strong> metastatic carcinoma <strong>of</strong> unknown origin: an algorithmicapproach. Semin Diagn Pathol 17:184–19330. Diaz Manzano JA, Medina Benegas A, Osete Alaladejo JM etal. (2003) Paraganglioma <strong>of</strong> <strong>the</strong> vagus. A case report <strong>and</strong> review<strong>of</strong> <strong>the</strong> literature. An Otorrinolaringol Ibero Am 30:127–13631. Dolgin SE, Groisman GM, Shah K (1995) Subcutaneousbronchogenic cysts <strong>and</strong> sinuses. Otolaryngol <strong>Head</strong> <strong>Neck</strong>Surg 112:763–766


<strong>Neck</strong> Cysts, Metastasis, Dissection Chapter 9 28132. Edmonds JL, Girod DA, Woodro<strong>of</strong> JM et al. (1997) Thirdbranchial anomalies. Avoiding recurrences. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 123:438–44133. Elmasalme F, Giacomantonio M , Clark KD et al. (2000)Congenital cervical teratoma in neonates, case report <strong>and</strong> review.Eur J Pediatr Surg 10:252–25734. Emery PJ, Baily CM, Evans JNG (1984) Cystic hygroma <strong>of</strong> <strong>the</strong>head <strong>and</strong> neck. A review <strong>of</strong> 37 cases. J Laryngol Otol 98:613–61935. Endicott JN, Cohen JJ (1979) Amyloidosis presenting as amass in <strong>the</strong> neck. Laryngoscope 89:1224–122836. Entwistle JWC, Pierce CV, Johnson DE et al. (1994) Parathyroidcyst: report <strong>of</strong> <strong>the</strong> sixth <strong>and</strong> youngest pediatric case. JPediatr Surg 29:1528–152937. Ettema SL, Caro<strong>the</strong>rs DG, H<strong>of</strong>fman HT (2003) Laryngoceleresection by combined external <strong>and</strong> endoscopic laser approach.Ann Otol Rhinol Laryngol 112:361–36438. Farm<strong>and</strong> M, Kuttenberger JJ (1996) A new <strong>the</strong>rapeutic conceptfor <strong>the</strong> treatment <strong>of</strong> cystic hygroma. Oral Surg Oral MedOral Pathol Oral Radiol Endod 81:389–39539. Fraga S, Helwig EB, Rosen SH (1971) Bronchogenic cysts in<strong>the</strong> skin <strong>and</strong> subcutaneous tissue. Am J Clin Pathol 56:230–23840. Franchi A, Meroni M, Massi D et al. (2002) Sinonasal undifferentiatedcarcinoma, nasopharyngeal-type undifferentiatedcarcinoma, <strong>and</strong> keratinizing <strong>and</strong> nonkeratinizing squamouscell carcinoma express differentiated cytokeratin patterns.Am J Surg Pathol 25:1597–160441. Frierson HF (1996) Cysts <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck sampled byfine-needle aspiration. Am J Clin Pathol 106:559–56042. Gillies EM, Luna MA (1998) Histologic evaluation <strong>of</strong> neckdissection specimens. Otolaryngol Clin North Am 31:759–77143. Golledge L, Ellis H (1994) The etiology <strong>of</strong> lateral cervical(branchial) cysts past, present <strong>the</strong>ories. Laryngol Otol108:653–65944. Gonzalez-Crussi F (1982) Extragonadal teratomas. In: Atlas<strong>of</strong> tumor pathology, 2nd edn. Fascicle 18. Armed Forces Institute<strong>of</strong> <strong>Pathology</strong>, Washington DC45. Grau C, Johansen LV, Jakobsen J et al. (2000). Cervical lymphnode metastases from unknown primary tumours. Resultsfrom a national survey by Danish Society for <strong>Head</strong> <strong>and</strong> <strong>Neck</strong>Oncology. Radio<strong>the</strong>r Oncol 55:121–12946. Gruber B, Rippon J, Dayal VS (1988) Phaeomycotic cyst(chromoblastomycosis) <strong>of</strong> <strong>the</strong> neck. Arch Otorhinolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 114:1031–103347. Guba Am, Adam AE, Jacques DA et al. (1978) Cervical presentation<strong>of</strong> thymic cysts. Am J Surg 130:430–43648. Hamy A, Masson S, Heymann MF et al. (2002) Parathyroidcyst. Report <strong>of</strong> 10 cases. Ann Chir 127:203–20749. Harney M, Pat IN, Walsh R et al. (2001) Laryngoceles <strong>and</strong>squamous cell carcinoma <strong>of</strong> <strong>the</strong> larynx. J Laryngol Otol115:590–59250. Heifetz SA, Cushings B, Giller R et al. (1998) Immature teratomain children: pathologic, considerations: a report from<strong>the</strong> combined Pediatric Oncology Group/Children’s CancerGroup. Am J Surg Pathol 22:1115–112451. Hendrickson M, Azarow K, Ein S et al. (1998) Congenitalthymic cysts in children mostly misdiagnosed. J Pediatr Surg33:821–82552. Hsieh YY, Hsueh S, Hsueh S et al. (2003) Pathologic analysis<strong>of</strong> congenital cervical cysts in children : 20 years experienceat Chang Gung Memorial Hospital. Chang Gung Med J26:107–11353. Iganej S, Kagan R, Anderson P et al. (2002) Metastatic squamouscell carcinoma <strong>of</strong> <strong>the</strong> neck from an unknown primary:management options <strong>and</strong> patterns <strong>of</strong> relapse. <strong>Head</strong> <strong>Neck</strong>24:236–24654. Jensen JL, Corell RW (1980) Nevus cells aggregates in subm<strong>and</strong>ibularlymph nodes. Oral Surg Oral Med Oral Pathol50:552–55655. Johnson TL, Zarbo RJ, Looyd RV et al. (1988) Paragangliomas<strong>of</strong> head <strong>and</strong> neck immunohistochemical neuroendocrine<strong>and</strong> intermediate filaments typing. Mod Pathol 1:216–22356. Jones D, Amin M, Ordonez NG (2001) Reticulum cell sarcoma<strong>of</strong> lymph nodes with mixed dendritic <strong>and</strong> fibroblastic features.Mod Pathol 14:1059–106757. Kenealy JF, Torsinglieri AR, Tom LW (1990) Branchial cleftanomalies: a five year retrospective study. Trans Pa AcadOphthalmol Otolaryngol 42:1022–102558. Khali RA, Prichep R, Minkowitz S (1984) Primary branchiogeniccarcinoma. <strong>Head</strong> <strong>Neck</strong> 11:153–15659. Koeller KK, Alamo L, Adair CF et al. (1999) Congenital cysticmasses <strong>of</strong> <strong>the</strong> neck: radiographic-pathologic correlation.Radiographics 19:121–14660. Kountakis SE, Minotti AM, Maillard A et al. (1994) Teratomas<strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Am J Otolaryngol 15:292–29661. Lack E, Cubilla AZ, Woodruff JM (1979) Paragangliomas <strong>of</strong><strong>the</strong> head <strong>and</strong> neck region: a pathologic study <strong>of</strong> tumors from73 patients. Hum Pathol 10:191–21862. Langman J (1975) Pharyngeal gut. In: Lagman J (eds) Medicalembryology, 3rd edn. Williams & Wilkins, Baltimore, pp260–28163. LaRouere MJ, Drake AF, Baker SR et al. (1987) Evaluation<strong>and</strong> management <strong>of</strong> a carcinoma arising in a thyroglossalduct cyst. Ann J Otolaryngol 8:351–35564. Lefebvre JL, Coche-Dequenant B, Ton Van J et al. (1990) Cervicallymph nodes from unknown primary tumor in 190 patients.Am J Surg 160:443–44665. Liu TP, Jeng KS, Yang TL et al. (1992) Thyroglossal duct cyst.An analysis <strong>of</strong> 92 cases. Clin Med J 49:72–7566. Luna MA, Pfaltz M (2000) Cysts <strong>of</strong> <strong>the</strong> neck, unknown primary<strong>and</strong> neck dissection. In: Gnepp D, (ed) Diagnostic surgicalpathology <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Saunders, Philadelphia,p 67167. Luna MA, Tortoledo ME, Allen M (1987) Salivary dermal analoguetumor arising in lymph nodes. Cancer 59:1165–116968. Maisel RH (1980) When your patient complains <strong>of</strong> a neckmass. Geriatrics 35:3–869. Martin H, Morfit HM, Ehrlich H (1950) The case for branchiogeniccancer (malignant branchioma). Ann Surg132:867–88770. May M (1978) <strong>Neck</strong> masses in children: diagnosis <strong>and</strong> treatment.Ear Nose Throat J 57:136–15871. Medina JE, Houck JR (1995) Advances in neck dissection.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 9:183–19672. Mendenhall WM, Mancuso AA, Parson J T et al. (1998) Diagnosticevaluation <strong>of</strong> squamous cell carcinoma metastaticto cervical lymph nodes from unknown head <strong>and</strong> neck primarysite. <strong>Head</strong> <strong>Neck</strong> 20:739–74473. Micheau C, Kijanienko J, Luboinski B et al. (1990) So-calledbranchiogenic carcinoma is actually cystic metastasis in <strong>the</strong>neck from a tonsillar primary. Laryngoscope 100:878–88374. Michelini ME, Casadio G, Franchella A (2003) Thyroglossalduct cysts: a retrospective study. Minerva Pediatr 55:51–5475. Molinari R, Cantu G, Chiesa F et al. (1977) A statistical approachto detection <strong>of</strong> <strong>the</strong> primary cancer based on <strong>the</strong> site <strong>of</strong>neck lymph node metastases. Tumori 63:267–28276. Moran C, Suster S, El-Naggar et al. (2004) Carcinoma arisingin multilocular thymic cysts <strong>of</strong> <strong>the</strong> neck: a clinicopathologicalstudy <strong>of</strong> three cases. Histopathology 44:64–6877. Mukherji SK, Drane WE, Mancuso AA et al. (1996) Occultprimary tumors <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck: detection with 2-[F-18] fluoro-2-deoxy-d-glucose SPECT. Radiology 199:761–76678. Mulliken JB, Glowacki J (1982) Hemangiomas <strong>and</strong> vascularmalformations in infants <strong>and</strong> children: a classification basedon endo<strong>the</strong>lial characteristics. Plastic Reconstr Surg 69:412–42079. Nguyen C. Shenouda G, Black MJ et al. (1994) Metastaticsquamous cell carcinoma to cervical lymph nodes from unknownprimary mucosal sites. <strong>Head</strong> <strong>Neck</strong> 16:58–6380. Nicollas R, Ducroz NE, Garabedian EN et al. (1998) Fourthbranchial pouch anomalies: a study <strong>of</strong> six cases <strong>and</strong> review <strong>of</strong><strong>the</strong> literature. Int J Pediatr Otorhinolaryngol 44:5–1081. Nozeran S, Duquenne M, Guyetan S et al. (2000) Diagnosis<strong>of</strong> parathyroid cysts: value <strong>of</strong> parathyroid hormone. PresseMed 29:939–941


282 M. A Luna · K. Pineda-Daboin982. Olsen KD, Maragos NE, Weil<strong>and</strong> LH (1980) First branchialcleft anomalies. Laryngoscope 90:423–43683. Ordonez NG (2000) Expression <strong>of</strong> thyroid transcription inhuman tumors. Adv Anat Pathol 7:120–12684. Paccioni D, Negro F, Valente G et al. (1994) Epstein-Barr virusby in situ hybridization in fine needle aspiration biopsy.Diagn Mol Pathol 3:100–10585. Papay FA, Kalucis C, Eliachar I et al. (1994) Nasopharyngealpresentation <strong>of</strong> a second branchial cleft cyst. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 110:232-23486. Park SS, Kamody CS (1992) The first branchial cleft carcinoma.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 118:969–97187. Peter JC, Sinclair-Smith C, de Villiers JC (1991) Midline dermalsinuses <strong>and</strong> cysts <strong>and</strong> <strong>the</strong>ir relationship to <strong>the</strong> centralnervous system. Eur J Pediatr Surg 1:73–7988. Pinczower E, Crocket DM, Atkinson JB et al. (1992) Preoperativethyroid scanning in presumed thyroglossal duct cysts.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 118:985–98889. Prasad KK, Gupta RK, Jain M et al. (2001) Cervical thymiccyst: report <strong>of</strong> a case <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Indian JPathol Microbiol 44:483–48590. Quick CA, Lowell SH (1977) Ranula <strong>and</strong> <strong>the</strong> sublingualgl<strong>and</strong>. Arch Otolaryngol 103:397–40091. Radkowski D, Arnold J, Healy GB et al. (1991) Thyroglossalduct remnants. Preoperative evaluation <strong>and</strong> management.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 117:1378–138192. Rangnekar H, Bailer WJ, Ghani A et al. (1996) Parathyroidcysts. Report <strong>of</strong> four cases <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. IntSurg 81:412–41493. Redaelli de Zinis LO, Nicolai P, Piccioni LO et al. (1998) Cervicalmetastatic carcinoma from an unknown primary: astudy on 65 patients. Br J Cancer 77S:2894. Regauer S, Beham A, Mannweiler MO (2000) CK7 expressionin carcinomas <strong>of</strong> <strong>the</strong> Waldeyer’s ring area. Hum Pathol31:1096–110195. Regauer S, Gogg-Kamerer, Braun H et al. (1997) Lateral neckcyst <strong>the</strong> branchial <strong>the</strong>ory revisited. A critical review <strong>and</strong>clinicopathologic study <strong>of</strong> 97 cases with special emphasis oncytokeratin expression. APMIS 105:623–63096. Regauer S, Manhweller S, Anderhuber A et al. (1999) Cysticlymph node metastases <strong>of</strong> squamous cell carcinoma <strong>of</strong>Waldeyer’s ring. Br J Cancer 79:1437–144297. Rhigi JD, S<strong>of</strong>ferman RA (1995) Screening unilateral tonsillectomyin <strong>the</strong> unknown primary Laryngoscope 105:548–55098. Richard JM, Micheau C (1997) Malignant cervical adenopathiesfrom carcinomas <strong>of</strong> unknown origin. Tumori 63:249–25899. Robbins KT, Clayman G, Levine PA et al. (2002) <strong>Neck</strong> dissectionclassification update. Revisions proposed by <strong>the</strong> American<strong>Head</strong> <strong>and</strong> <strong>Neck</strong> Society <strong>and</strong> <strong>the</strong> American Academy <strong>of</strong>Otolaryngology—<strong>Head</strong> <strong>and</strong> <strong>Neck</strong> Surgery. Arch Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 128:751–758100. Rodriguez-Cuevas S, Lopez-Garza J, Labastida-Almendaro S(1998) Carotid body tumors in inhabitants <strong>of</strong> altitudes higherthan 2000 meters above sea level. <strong>Head</strong> <strong>Neck</strong> 20:374–37101. Rosen D, Wirtschafer A, Rao VM et al. (1998) Dermoid cyst<strong>of</strong> <strong>the</strong> lateral neck, a case report <strong>and</strong> literature review. EarNose Throat J 125:129–132102. Sade J, Rosen G (1968) Thyroglossal cysts <strong>and</strong> tracts: a histological<strong>and</strong> histochemical study. Ann Otol Rhino Laryngo177:139–145103. Saenz J, Catalina F, Fernández MJJ et al. (2003) Mucoepidermoidcarcinoma arising in cervical lymph nodes. A report <strong>of</strong>three cases with FNA findings. Acta Cytol 47:470–474104. Saitz EW (1981) Cervical lymphadenitis caused by atypicalmycobacteria. Pediatr Clin North Am 28:823–839105. Sambola-Cabrer I, Fern<strong>and</strong>ez-Real JM, Ricard W (1996) Ectopicthyroid tissue presenting as a subm<strong>and</strong>ibular mass.<strong>Head</strong> <strong>Neck</strong> 18:87–90106. Samuel M, Freeman NV, Sajwany MJ (1993) Lingual thyroglossalduct cyst presenting in infancy 28:891–893107. Scherer K, Szeimies RM, L<strong>and</strong>thaler M (2000) Ectopic parotidtissue. An unusual differential cervical cystic tumor diagnosis.Hautarzt 51:865–868108. Senger MI, Applebaum EL, Ley KD (1979) Heterotopic salivarytissue in <strong>the</strong> neck. Laryngoscope 80:1772–1778109. Shah JP (1990) Patterns <strong>of</strong> cervical lymph node metastasisfrom squamous cell carcinoma <strong>of</strong> <strong>the</strong> upper aerodigestivetract. Am J Surg 160:405–409110. Shah JP, Strang E, Spiro RH et al. (1981) <strong>Neck</strong> dissection, currentstatus <strong>and</strong> future possibilities. Clin Bull 11:25–33111. Shvero J, Hadar TA, Vidos I et al. (1986) Heterotopic salivarytissue <strong>and</strong> branchial sinuses. Laryngol Otol 100:243–246112. Sistrunk WE (1920) The surgical treatment <strong>of</strong> cysts <strong>of</strong> <strong>the</strong>thyroglossal tract. Ann Surg 71:121–122113. Smirniotopoulos JG, Chiechi MV (1995) Teratomas, dermoids,<strong>and</strong> epidermoids <strong>of</strong> <strong>the</strong> head <strong>and</strong> neck. Radiographics15:1437–1455114. Soyla L, Aydogan LB, Kiroglu M et al. (1995) Hydatidic cystin <strong>the</strong> head <strong>and</strong> neck area. Am J Otolaryngol 16:123–125115. Spinelli C, Ricci E, Berti P et al. (1990) <strong>Neck</strong> mass in childhood.Surgical experience <strong>of</strong> 154 cases. Minerva Pediatr42:169–172116. Thompson L, Heffner DK (1998) The clinical importance <strong>of</strong>cystic squamous cell carcinoma in <strong>the</strong> neck. Cancer 82:944–956117. Torsiglier AJ Jr, Tom LW, Ross AJ et al. (1988) Pediatric neckmasses: guidelines for evaluation. Int Pediatr Otorhinolaryngol16:199–210118. Tovi F, Fliss DM, Inbar-Yanai I (1988) Hürthle cell adenoma<strong>of</strong> <strong>the</strong> thyroglossal duct. <strong>Head</strong> <strong>Neck</strong> Surg 10:346–349119. Triglia JM, Nicollas R, Ducroz V et al. (1998) First branchialcleft anomalies. A study <strong>of</strong> 39 cases <strong>and</strong> a review <strong>of</strong> <strong>the</strong> literature.Arch Otolaryngol <strong>Head</strong> <strong>Neck</strong> Surg 124:291–295120. Turner A, Lampe FB, Cramer H (1989) Parathyroid cysts. JOtolaryngol 18:11–13121. Tyson RW, Gr<strong>of</strong>f DB (1993) An unusual lateral neck cyst with<strong>the</strong> combined features <strong>of</strong> a bronchogenic, thyroglossal, <strong>and</strong>branchial cleft origin. Pediatr Pathol 13:567–572122. Vermeire VM, Dale JJ (1991) Second branchial cleft-pouchset fistulae, sinuses <strong>and</strong> cysts in children. Acta OtorhinolaryngolBelg 45:437–442123. Wang RC, Goepfert H, Barber AE et al. (1990) Unknown primarySCC metastatic to <strong>the</strong> neck. Arch Otolaryngol <strong>Head</strong><strong>Neck</strong> Surg 116:1388–1393124. Wassef M (1992) Cervicophalic hemangiomas <strong>and</strong> vascularmalformations. Histological appearance <strong>and</strong> classification. JMal Vasc 17:20–25125. Werner AJ, Dunne AA, Myers JN (2003) Functional anatomy<strong>of</strong> <strong>the</strong> lymphatic drainage system <strong>of</strong> <strong>the</strong> upper aerodigestivetract <strong>and</strong> its role in metastasis <strong>of</strong> squamous cell carcinoma.<strong>Head</strong> <strong>Neck</strong> 25:322–332126. Wilson DB (1979) Embryonic development <strong>of</strong> <strong>the</strong> head <strong>and</strong>neck. II. The branchial region. <strong>Head</strong> <strong>Neck</strong> Surg 2:59–66127. Yang C, Cohen J, Everts E et al. (1999) Fourth branchial archsinus: clinical presentation, diagnostic workup, <strong>and</strong> surgicaltreatment. Laryngoscope 109:442–446128. Yoo KS, Chengazi VU, O’Mara RE (1998) Thyroglossal ductcyst with papillary carcinoma in a 11-year-old girl. J PediatrSurg 33:745–746129. Yoshimura Y, Obara S, Kondoh T et al. (1995) A comparison<strong>of</strong> three methods used for treatment <strong>of</strong> ranula. J Oral Maxoill<strong>of</strong>acSurg 53:280–282130. Zarbo RJ, McClatchey KD, Arcen RG et al. (1983) Thymopharyngealduct cyst; a form <strong>of</strong> cervical thymus. Ann OtolRhinol Laryngol 92:284–289131. Zatchuz JT, Paton CA, Hyams VJ (1982) Cervical heterotopicsalivary gl<strong>and</strong> neoplasms: a diagnostic dilemma. Otolaryngol<strong>Head</strong> <strong>Neck</strong> Surg 80:178–181132. Zimmermann CE, Domarus H von, Moubayed P (2002) Carcinomain situ in a lateral cervical cyst. <strong>Head</strong> <strong>Neck</strong> 24:965–969


Chapter 10Eye<strong>and</strong> Ocular AdnexaM.R. Canninga-Van Dijk10Contents10.1 Summary <strong>of</strong> Anatomy <strong>and</strong> Histology . . . . . . . . . 28410.1.1 Conjunctiva . . . . . . . . . . . . . . . . . . . . . . . 28410.1.2 Cornea . . . . . . . . . . . . . . . . . . . . . . . . . . 28410.1.3 Intraocular Tissues . . . . . . . . . . . . . . . . . . . 28410.1.4 Optic Nerve . . . . . . . . . . . . . . . . . . . . . . . 28410.1.5 Lacrimal Gl<strong>and</strong>s <strong>and</strong> Lacrimal Passages . . . . . . . 28410.1.6 Eyelids . . . . . . . . . . . . . . . . . . . . . . . . . . 28410.1.7 Orbit . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28510.2 Conjunctiva . . . . . . . . . . . . . . . . . . . . . . . 28510.2.1 Developmental Anomalies . . . . . . . . . . . . . . . 28510.2.1.1 Dermoid, Dermolipoma <strong>and</strong> Complex Choristoma 28510.2.2 Cysts . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28510.2.2.1 Inclusion cysts . . . . . . . . . . . . . . . . . . . . . . 28510.2.3 Degeneration . . . . . . . . . . . . . . . . . . . . . . . 28610.2.3.1 Pinguecula <strong>and</strong> Pterygium . . . . . . . . . . . . . . . 28610.2.4 Inflammatory Processes . . . . . . . . . . . . . . . . 28610.2.4.1 Acute Conjunctivitis . . . . . . . . . . . . . . . . . . 28610.2.4.2 Chronic Non-Granulomatous Conjunctivitis . . . . 28710.2.4.3 Granulomatous Conjunctivitis . . . . . . . . . . . . 28710.2.4.4 Ligneous Conjunctivitis . . . . . . . . . . . . . . . . 28710.2.4.5 Chlamydia Trachomatis (TRIC Agent) Infection . . 28710.2.5 Dermatologic <strong>and</strong> Systemic Diseases . . . . . . . . . 28810.2.5.1 Keratoconjunctivitis Sicca . . . . . . . . . . . . . . . 28810.2.5.2 Dermatologic Diseases . . . . . . . . . . . . . . . . . 28810.2.5.3 Metabolic Diseases . . . . . . . . . . . . . . . . . . . 28810.2.6 Tumours <strong>and</strong> Tumour-Like Conditions . . . . . . . 28810.2.6.1 Epi<strong>the</strong>lial . . . . . . . . . . . . . . . . . . . . . . . . . 28810.2.6.2 Melanocytic . . . . . . . . . . . . . . . . . . . . . . . 29010.2.6.3 O<strong>the</strong>r Neoplasms . . . . . . . . . . . . . . . . . . . . 29110.3 Cornea . . . . . . . . . . . . . . . . . . . . . . . . . . 29210.3.1 Keratitis <strong>and</strong> Corneal Ulcers . . . . . . . . . . . . . . 29210.3.1.1 Herpes Simplex Keratitis . . . . . . . . . . . . . . . . 29210.3.1.2 Corneal Ulceration Due to Systemic Disease . . . . 29210.3.2 Keratoconus . . . . . . . . . . . . . . . . . . . . . . . 29210.3.3 Hereditary Corneal Dystrophies . . . . . . . . . . . 29310.3.3.1 Epi<strong>the</strong>lial Dystrophies . . . . . . . . . . . . . . . . . 29310.3.3.2 Stromal Dystrophies . . . . . . . . . . . . . . . . . . 29310.3.3.3 Endo<strong>the</strong>lial Dystrophies . . . . . . . . . . . . . . . . 29310.3.4 Failed Previous Grafts . . . . . . . . . . . . . . . . . 29410.4 Intraocular Tissues . . . . . . . . . . . . . . . . . . . 29410.4.1 Developmental Anomalies . . . . . . . . . . . . . . . 29410.4.1.1 Congenital Glaucoma . . . . . . . . . . . . . . . . . . 29410.4.1.2 Retinopathy <strong>of</strong> Prematurity . . . . . . . . . . . . . . 29410.4.1.3 Persistent Primary Hyperplastic Vitreous . . . . . . 29410.4.1.4 Retinal Dysplasia . . . . . . . . . . . . . . . . . . . . 29410.4.1.5 Aniridia . . . . . . . . . . . . . . . . . . . . . . . . . . 29410.4.1.6 Congenital Rubella Syndrome . . . . . . . . . . . . . 29410.4.2 Inflammatory Processes . . . . . . . . . . . . . . . . 29510.4.2.1 Acute Inflammation . . . . . . . . . . . . . . . . . . 29510.4.2.2 Chronic Non-Granulomatous Inflammation . . . . 29510.4.2.3 Granulomatous Inflammation . . . . . . . . . . . . . 29510.4.3 Trauma . . . . . . . . . . . . . . . . . . . . . . . . . . 29610.4.4 Degeneration . . . . . . . . . . . . . . . . . . . . . . . 29610.4.4.1 Glaucoma . . . . . . . . . . . . . . . . . . . . . . . . . 29610.4.4.2 Cataracts . . . . . . . . . . . . . . . . . . . . . . . . . 29710.4.4.3 Phtisis Bulbi . . . . . . . . . . . . . . . . . . . . . . . 29810.4.4.4 Retinal Vascular Disease . . . . . . . . . . . . . . . . 29810.4.4.5 Retinal Detachment . . . . . . . . . . . . . . . . . . . 29810.4.4.6 Retinitis Pigmentosa . . . . . . . . . . . . . . . . . . 29810.4.5 Tumours <strong>and</strong> Tumour-Like Conditions . . . . . . . 29810.4.5.1 Melanocytic . . . . . . . . . . . . . . . . . . . . . . . 29810.4.5.2 Lymphoid . . . . . . . . . . . . . . . . . . . . . . . . . 30010.4.5.3 Retinoblastoma <strong>and</strong> Pseudoretinoblastoma . . . . . 30010.4.5.4 Glial . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30110.4.5.5 Vascular . . . . . . . . . . . . . . . . . . . . . . . . . 30110.4.5.6 O<strong>the</strong>r Primary Tumours . . . . . . . . . . . . . . . . 30110.4.5.7 Metastatic Tumours . . . . . . . . . . . . . . . . . . . 30210.5 Optic Nerve . . . . . . . . . . . . . . . . . . . . . . . 30210.5.1 Papilloedema . . . . . . . . . . . . . . . . . . . . . . . 30210.5.2 Optic Neuritis . . . . . . . . . . . . . . . . . . . . . . 30210.5.3 Optic Atrophy . . . . . . . . . . . . . . . . . . . . . . 30210.5.4 Tumours . . . . . . . . . . . . . . . . . . . . . . . . . 30210.5.4.1 Glioma . . . . . . . . . . . . . . . . . . . . . . . . . . 30210.5.4.2 Meningioma . . . . . . . . . . . . . . . . . . . . . . . 30210.6 Lacrimal Gl<strong>and</strong> <strong>and</strong> Lacrimal Passages . . . . . . . 30210.6.1 Inflammatory Processes . . . . . . . . . . . . . . . . 30210.6.2 Tumours <strong>and</strong> Tumour-Like Conditions . . . . . . . 30310.7 Eyelids . . . . . . . . . . . . . . . . . . . . . . . . . . 30310.7.1 Cysts . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30310.7.1.1 Dermoid Cyst . . . . . . . . . . . . . . . . . . . . . . 30310.7.1.2 Epidermal Cyst . . . . . . . . . . . . . . . . . . . . . 30310.7.1.3 Hidrocystoma . . . . . . . . . . . . . . . . . . . . . . 30310.7.2 Inflammatory Processes . . . . . . . . . . . . . . . . 30310.7.2.1 Chalazion <strong>and</strong> O<strong>the</strong>r Ruptured Cysts . . . . . . . . 30410.7.2.2 Deep Granuloma Annulare . . . . . . . . . . . . . . 30410.7.2.3 Necrobiotic Xanthogranuloma . . . . . . . . . . . . 30410.7.3 Amyloidosis . . . . . . . . . . . . . . . . . . . . . . . 30510.7.4 Tumours <strong>and</strong> Tumour-Like Conditions . . . . . . . 30510.7.4.1 Xan<strong>the</strong>lasmata . . . . . . . . . . . . . . . . . . . . . . 30510.8 Orbit . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30510.8.1 Inflammatory Processes . . . . . . . . . . . . . . . . 30510.8.1.1 Dysthyroid Ophthalmopathy . . . . . . . . . . . . . 30510.8.1.2 Cellulitis . . . . . . . . . . . . . . . . . . . . . . . . . 30510.8.1.3 Pseudotumour . . . . . . . . . . . . . . . . . . . . . . 30610.8.2 Tumours <strong>and</strong> Tumour-Like Conditions . . . . . . . 30610.8.2.1 Developmental Cysts . . . . . . . . . . . . . . . . . . 30610.8.2.2 Optic Nerve <strong>and</strong> Meningeal Tumours . . . . . . . . 30610.8.2.3 Metastatic Tumours . . . . . . . . . . . . . . . . . . . 307References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307


284 M.R. Canninga-Van Dijk1010.1 Summary <strong>of</strong> Anatomy<strong>and</strong> Histology10.1.1 ConjunctivaThe conjunctiva is a thin mucous membrane, coveringmost <strong>of</strong> <strong>the</strong> anterior surface <strong>of</strong> <strong>the</strong> eye <strong>and</strong> <strong>the</strong> innersurface <strong>of</strong> <strong>the</strong> eyelids. The conjunctiva is composed<strong>of</strong> two to five layers <strong>of</strong> stratified columnar epi<strong>the</strong>liumcontaining mucin-secreting goblet cells. At <strong>the</strong> corneoscleraljunction, <strong>the</strong> limbus, a gradual transition fromstratified columnar to stratified squamous cornealepi<strong>the</strong>lium is seen. At <strong>the</strong> palpebral margins, a relativelyabrupt transition into <strong>the</strong> epidermis is present.The basal layer contains melanocytes <strong>and</strong> Langerhanscells. The conjunctival stroma is composed <strong>of</strong> fibrousconnective tissue containing fibroblasts, some inflammatorycells, blood vessels, smooth muscle, nerves <strong>and</strong>lymphatic channels. In <strong>the</strong> fornices <strong>the</strong> conjunctivalepi<strong>the</strong>lium contains more goblet cells <strong>and</strong> <strong>the</strong> stromacan additionally contain cartilage. In <strong>the</strong> medial interpalpebralarea <strong>of</strong> <strong>the</strong> eye, a nodular mass is present: <strong>the</strong>caruncle. The caruncle is covered by stratified non-keratinisedsquamous epi<strong>the</strong>lium <strong>and</strong> <strong>the</strong> subepi<strong>the</strong>lialstroma contains sebaceous gl<strong>and</strong>s, hair follicles <strong>and</strong>muscle fibres.10.1.2 CorneaThe cornea is divisible into five distinctive layers: <strong>the</strong>epi<strong>the</strong>lium, Bowman’s layer, <strong>the</strong> stroma, Descemet’smembrane <strong>and</strong> <strong>the</strong> endo<strong>the</strong>lium. The non-keratinisingstratified squamous epi<strong>the</strong>lium consists in <strong>the</strong> centre <strong>of</strong><strong>the</strong> cornea <strong>of</strong> five layers, increasing to nine or ten layersat <strong>the</strong> periphery. Bowman’s layer is believed to representa modified layer <strong>of</strong> <strong>the</strong> stroma. It is composed <strong>of</strong> smallcollagen fibrils surrounded by a mucoprotein groundsubstance. The stroma is avascular <strong>and</strong> consists <strong>of</strong> collagenouslamellae arranged in an almost parallel mannerinterspersed with flattened fibroblasts (keratocytes).Descemet’s membrane is a faintly eosinophilic staining,PAS-positive, acellular structure, formed by corneal endo<strong>the</strong>lialcells. The underlying endo<strong>the</strong>lium is a singlelayer <strong>of</strong> polygonal cells.10.1.3 Intraocular TissuesThe intra-ocular tissue is formed by <strong>the</strong> uveal tract,<strong>the</strong> retina, <strong>the</strong> lens <strong>and</strong> <strong>the</strong> vitreous gel. The uveal tractconsists <strong>of</strong> highly vascularised, loosely arranged stroma,which can be found in all three parts <strong>of</strong> <strong>the</strong> tract:<strong>the</strong> iris, <strong>the</strong> ciliary body <strong>and</strong> <strong>the</strong> choroid. In <strong>the</strong> iris,in addition to <strong>the</strong> stroma, <strong>the</strong> pigment epi<strong>the</strong>lium <strong>and</strong><strong>the</strong> smooth muscles <strong>of</strong> <strong>the</strong> musculus sphincter pupillae<strong>and</strong> musculus dilator pupillae can be found. The ciliarybody consists <strong>of</strong> two parts: <strong>the</strong> pars plicata contains<strong>the</strong> ciliary processes, <strong>the</strong> pars plana serves as an attachmentfor <strong>the</strong> vitreous gel. In both parts, layers <strong>of</strong> ciliarymuscle are present. The choroid contains nerves <strong>and</strong> avariable number <strong>of</strong> melanocytes, related to race. Theretina consists <strong>of</strong> a nerve fibre layer, peripheral ganglioncells, <strong>the</strong> bipolar cell layer, <strong>the</strong> photoreceptor cells<strong>and</strong> <strong>the</strong> retinal pigment epi<strong>the</strong>lium. The thickness <strong>of</strong><strong>the</strong> retina varies in different regions. The biconvex lensis a transparent structure, <strong>the</strong> form <strong>of</strong> which is easilyaltered by contraction <strong>of</strong> <strong>the</strong> ciliary muscles. The shape<strong>of</strong> <strong>the</strong> lens is maintained by <strong>the</strong> elastic lens capsule. Ina fixed specimen <strong>the</strong> lens is rigid, caused by coagulation<strong>of</strong> <strong>the</strong> soluble crystallins. The inner surface <strong>of</strong> <strong>the</strong>anterior lens capsule is covered with a single layer <strong>of</strong>cuboidal epi<strong>the</strong>lium. A loose framework <strong>of</strong> type II collagencontaining mainly water <strong>and</strong> acid mucopolysaccharidesforms <strong>the</strong> vitreous gel.10.1.4 Optic NerveThe axons from <strong>the</strong> retinal nerve fibre layer converge at<strong>the</strong> optic disc. The axons in <strong>the</strong> nerve fibre layer <strong>of</strong> <strong>the</strong>optic disc form a bulge as <strong>the</strong>y pass through <strong>the</strong> laminacribrosa, a sieve-like plate <strong>of</strong> connective tissue formedby fibroblasts, ingrowing from <strong>the</strong> posterior part <strong>of</strong> <strong>the</strong>adjacent sclera. This bulge is larger on <strong>the</strong> nasal side.The tissue anterior to <strong>the</strong> lamina derives blood supplyfrom <strong>the</strong> posterior ciliary arteries, <strong>and</strong> <strong>the</strong> tissueposterior to <strong>the</strong> lamina has a meningeal blood supplyderived from branches <strong>of</strong> <strong>the</strong> ophthalmic <strong>and</strong> centralretinal artery.10.1.5 Lacrimal Gl<strong>and</strong>s<strong>and</strong> Lacrimal PassagesThe pale brown <strong>and</strong> ovoid lacrimal gl<strong>and</strong> is located in<strong>the</strong> upper outer orbit. The large ducts pass through <strong>the</strong>conjunctival epi<strong>the</strong>lium into <strong>the</strong> superior fornix. Thetear fluids drain into <strong>the</strong> canaliculi, ending in <strong>the</strong> lacrimalsac. The secretory gl<strong>and</strong> is composed <strong>of</strong> lobulargrouped acini, secreting solutes <strong>and</strong> glycosaminoglycans.The drainage system consists <strong>of</strong> ductules, formedby epi<strong>the</strong>lial cells surrounded by myoepi<strong>the</strong>lium. Thecanaliculi are covered with stratified epi<strong>the</strong>lium, while<strong>the</strong> lacrimal sac is covered with columnar epi<strong>the</strong>lium.10.1.6 EyelidsThe eyelids are covered by an epidermis above a thindermis with small sweat gl<strong>and</strong>s <strong>and</strong> pilosebaceous units.


Eye <strong>and</strong> Ocular Adnexa Chapter 10 285The dermis covers <strong>the</strong> musculus orbicularis oculi, locatedon <strong>the</strong> anterior surface <strong>of</strong> <strong>the</strong> tarsal plate, which iscomposed <strong>of</strong> compact stroma. In <strong>the</strong> anterior part <strong>of</strong> <strong>the</strong>eyelids, <strong>the</strong> pilosebaceous units are much larger to form<strong>the</strong> lashes. The parts <strong>of</strong> <strong>the</strong> tarsal plates closest to <strong>the</strong>lid margins, contain <strong>the</strong> large sebaceous (Meibomian)gl<strong>and</strong>s. A transition from keratinised squamous epi<strong>the</strong>lium<strong>of</strong> <strong>the</strong> outer eyelid into columnar epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong>tarsal conjunctiva is present in <strong>the</strong> transition zone. Smallpilosebaceous gl<strong>and</strong>s (Zeis) <strong>and</strong> sweat gl<strong>and</strong>s (Moll) arepresent at <strong>the</strong> lid margins.10.1.7 OrbitThe orbital septum <strong>and</strong> globe divide <strong>the</strong> orbit intoanterior <strong>and</strong> posterior compartments. The anteriorcompartment consists <strong>of</strong> <strong>the</strong> lids, lacrimal apparatus<strong>and</strong> anterior s<strong>of</strong>t tissues. The posterior compartmentis also called <strong>the</strong> retrobulbar space. The cone <strong>of</strong> thisretrobulbar space consists <strong>of</strong> <strong>the</strong> extraocular muscles<strong>and</strong> an envelope <strong>of</strong> fascia. The optic nerve is locatedwithin <strong>the</strong> intraconal space, surrounded by fibrous <strong>and</strong>fatty tissue.Fig. 10.1. Dermolipoma: excision <strong>of</strong> a limbal mass, consisting <strong>of</strong>collagen <strong>and</strong> adipose tissue10.2 ConjunctivaBecause conjunctival biopsies <strong>and</strong> excisions are verythin, <strong>the</strong>y tend to fold when placed into fixative. Withoutcausing compression artefacts, <strong>the</strong> surgeon shouldspread <strong>the</strong> tissue onto a piece <strong>of</strong> cardboard or filter paper<strong>and</strong> let it dry for a few seconds, before placing it into <strong>the</strong>fixative.10.2.1 Developmental AnomaliesFig. 10.2. Dermolipoma: detail <strong>of</strong> <strong>the</strong> stratified squamous epi<strong>the</strong>liumcontaining hair follicles10.2.1.1 Dermoid, Dermolipoma<strong>and</strong> Complex ChoristomaICD-O:9084/0Dermoid tumours are firm or cystic masses, typically occurringat <strong>the</strong> limbus on <strong>the</strong> temporal side <strong>and</strong> are <strong>the</strong>most frequent epibulbar tumours in children [30]. Theyare composed <strong>of</strong> epi<strong>the</strong>lial <strong>and</strong>/or connective tissue elementsthat become entrapped within embryonic clefts. Onhistological examination, <strong>the</strong> surface epi<strong>the</strong>lium consists<strong>of</strong> stratified squamous epi<strong>the</strong>lium, frequently containingskin appendages. The stromal component consists <strong>of</strong> collagenarranged in thick bundles; it contains blood vessels<strong>and</strong> nerve fibres. If adipose tissue is present, <strong>the</strong> lesion iscalled dermolipoma (Figs. 10.1, 10.2). Sometimes cartilageor lacrimal gl<strong>and</strong> tissue is present, <strong>the</strong>se lesions areknown as complex choristoma [28, 35].Dermoids are present at birth <strong>and</strong> have little or nogrowth potential. The lesions can occur as isolated ocularlesions or in association with anomalies affectingo<strong>the</strong>r organs (Goldenhar’s syndrome, m<strong>and</strong>ibul<strong>of</strong>acialdysostosis <strong>and</strong> neurocutaneous syndrome) [22, 41, 46,55, 100, 118].10.2.2 Cysts10.2.2.1 Inclusion CystsConjunctival inclusion cysts are acquired lesions, usuallyarising following surgical or accidental trauma [9, 42,83]. The cysts are lined with non-keratinising cuboidal


286 M.R. Canninga-Van Dijkepi<strong>the</strong>lium with apocrine changes <strong>and</strong> containing gobletcells (Fig. 10.3). In <strong>the</strong> underlying stroma, chronic inflammatorycells may be present.10.2.3 Degeneration10.2.3.1 Pinguecula<strong>and</strong> Pterygium10Pingueculae are raised, localised, yellowish-grey lesionsthat occur in <strong>the</strong> bulbar conjunctiva, close to <strong>the</strong> limbuson <strong>the</strong> nasal or temporal side <strong>of</strong> <strong>the</strong> cornea. Pterygiaare similar in appearance <strong>and</strong> also develop in <strong>the</strong>se areas,but involve <strong>the</strong> peripheral cornea, mostly <strong>the</strong> nasalside, as well. Pingueculae <strong>and</strong> pterygia are degenerativelesions causally related to prolonged actinic exposure.The lesions are <strong>of</strong>ten bilateral <strong>and</strong> occur in middleaged<strong>and</strong> elderly patients, especially in areas with highlevels <strong>of</strong> sunlight. On histologic examination both lesionsare identical. The essential feature is elastotic degeneration<strong>of</strong> <strong>the</strong> collagen, resulting in a subepi<strong>the</strong>lialzone <strong>of</strong> amorphous, basophilic material (Fig. 10.4). Thismaterial stains black with <strong>the</strong> Elastica van Gieson stain(Fig. 10.5). In older lesions calcification can occur. Theoverlying epi<strong>the</strong>lium may show a wide variety <strong>of</strong> changes,but most frequently it is thin, atrophic conjunctivalepi<strong>the</strong>lium or acanthosis without cellular atypia. In <strong>the</strong>epi<strong>the</strong>lium an actinic keratosis or even a squamous cellcarcinoma may develop [110].Since pingueculae are not progressive, <strong>the</strong>y are seldomexcised. Pterygia are <strong>of</strong> more clinical importancebecause <strong>of</strong> <strong>the</strong>ir extension to <strong>the</strong> cornea.10.2.4 Inflammatory ProcessesThe conjunctiva is prone to inflammations with manydifferent causes, ei<strong>the</strong>r infectious or as a part <strong>of</strong> a noninfectiousdermatologic or systemic disease. Usually<strong>the</strong>se lesions do not cause diagnostic problems, but abiopsy can be useful in making <strong>the</strong> correct diagnosis[97]. In this chapter <strong>the</strong> inflammatory processes <strong>of</strong> <strong>the</strong>conjunctiva are divided into acute, chronic <strong>and</strong> granulomatous.The only lesions mentioned separately are ligneousconjunctivitis <strong>and</strong> lesions caused by Chlamydiainfections.Fig. 10.3. Conjunctival inclusion cyst: a cystic space is covered withnon-keratinising cuboidal epi<strong>the</strong>lium, containing goblet cellsFig. 10.4. Pinguecula: amorphous, basophilic material in <strong>the</strong>stroma represents <strong>the</strong> elastotic degenerated collagen10.2.4.1 Acute ConjunctivitisIn acute conjunctivitis <strong>the</strong>re is a rapid onset <strong>of</strong> a swollen,hyperaemic conjunctiva accompanied by increasedtear formation. There may be a watery (viral infections),fibrinous (bacterial infections) or mucoid (allergic reac-Fig. 10.5. Pinguecula: <strong>the</strong> elastotic collagen stains black in elasticastaining (Elastica van Gieson stain)


Eye <strong>and</strong> Ocular Adnexa Chapter 10 287tions) discharge [124]. The histological pattern dependson <strong>the</strong> cause <strong>of</strong> <strong>the</strong> inflammation. In viral infections<strong>the</strong> infiltrate consists mostly <strong>of</strong> mononuclear cells. In abacterial infection many neutrophilic leukocytes can beseen [124]. When <strong>the</strong> cause is an allergic response, manyeosinophils are usually found.10.2.4.2 Chronic Non-GranulomatousConjunctivitisChronic conjunctivitis can be caused by many infectious,immunological <strong>and</strong> toxic agents. Also, anatomic aberrations(like ectropion or proptosis) can cause inflammation.In chronic conjunctivitis <strong>the</strong> epi<strong>the</strong>lium becomeshyperplastic <strong>and</strong> <strong>the</strong> goblet cells increase in number.Crypt-like epi<strong>the</strong>lial infoldings can occur, forming subepi<strong>the</strong>lialretention cysts. These cysts contain mucus inwhich calcification can be seen over time. The presence<strong>of</strong> perivascular infiltrate in <strong>the</strong> stroma can induce fibrousb<strong>and</strong>s between <strong>the</strong> epi<strong>the</strong>lium <strong>and</strong> <strong>the</strong> tarsus, which cancause surface irregularities, <strong>the</strong> so-called papillary conjunctivitis.In fact, <strong>the</strong> epi<strong>the</strong>lial <strong>and</strong> stromal responses <strong>of</strong>a papillary conjunctivitis are non-specific <strong>and</strong> can also beseen in atopic conjunctivitis <strong>and</strong>, in a more extreme formin individuals wearing contact lenses (giant papillary conjunctivitis)[108, 114]. When lymph follicles are found in<strong>the</strong> superficial stroma, it is called follicular conjunctivitis.The presence <strong>of</strong> <strong>the</strong>se follicles is associated with adenoviralinfections [25]. It can also be seen in early chlamydialinfections [37], Borrelia burgdorferi infections [60, 132]<strong>and</strong> in patients using topical medication. In <strong>the</strong>se situationsa lymphoma has to be excluded by immunohistochemistry[2, 48, 131].With long-st<strong>and</strong>ing inflammation <strong>the</strong> epi<strong>the</strong>liumcan become atrophic, with loss <strong>of</strong> goblet cells. The epi<strong>the</strong>liumcan show keratinisation, resulting in a white appearance(leukoplakia). The long-st<strong>and</strong>ing inflammationmay result in scarring <strong>of</strong> <strong>the</strong> conjunctival stroma.10.2.4.3 Granulomatous ConjunctivitisGranulomatous inflammation <strong>of</strong> <strong>the</strong> conjunctiva is usuallyassociated with systemic diseases. The presence <strong>of</strong>exogenous material or parasites may cause an isolatedgranulomatous inflammation <strong>of</strong> <strong>the</strong> conjunctiva. In <strong>the</strong>case <strong>of</strong> a chalazion <strong>the</strong> palpebral conjunctiva may alsobe affected (see Sect. 10.7.2.1).10.2.4.3.1 InfectiousLike in o<strong>the</strong>r anatomic sites, a caseating necrotisinggranulomatous infection can be caused by tuberculosis,especially in children [32, 45, 76].10.2.4.3.2 SarcoidosisIn sarcoidosis <strong>the</strong> granulomas are usually small <strong>and</strong>sharply demarcated [6, 39]. Serial sections can be necessaryto confirm <strong>the</strong> diagnosis.10.2.4.4 Ligneous ConjunctivitisLigneous conjunctivitis (chronic pseudomembranousconjunctivitis) is a rare bilateral disease, mainly occurringin young girls. It presents as a subacute inflammation<strong>of</strong> <strong>the</strong> tarsal conjunctiva, <strong>of</strong>ten accompanied bynasopharyngitis <strong>and</strong> vaginitis. The disease seems to bedue to a defective fibrinolysin system. Microscopically,granulation tissue is present, covered with plaques <strong>of</strong>fibrinous material, later forming a hyalinised mass. Afterremoval <strong>of</strong> <strong>the</strong> plaque, recurrence is common. It canbe complicated by corneal involvement <strong>and</strong> perforationwith loss <strong>of</strong> <strong>the</strong> eye may occur [15, 17, 99].10.2.4.5 Chlamydia Trachomatis(TRIC Agent) InfectionChlamydia trachomatis (TRIC agent: TR = trachoma,IC = inclusion conjunctivitis) is an obligate intracellularpathogen <strong>of</strong> columnar epi<strong>the</strong>lial cells [79]. In hotclimates it can cause trachoma, primarily affecting <strong>the</strong>conjunctiva <strong>and</strong> corneal epi<strong>the</strong>lium, ultimately causingcicatrisation <strong>of</strong> this tissue. Trachoma commonlyaffects children <strong>and</strong> is one <strong>of</strong> <strong>the</strong> world’s major causes<strong>of</strong> blindness. Trachoma is spread from eye to eye bytransfer <strong>of</strong> ocular discharges. In temperate climateschlamydial infection is venereal, with only mild conjunctivalinfection, called inclusion conjunctivitis. Athird manifestation <strong>of</strong> this pathogen is ophthalmianeonatorum [75].10.2.4.5.1 TrachomaThe clinical manifestations <strong>of</strong> trachoma vary with<strong>the</strong> severity <strong>and</strong> duration <strong>of</strong> <strong>the</strong> initial infection <strong>and</strong>also depend upon environmental factors, <strong>the</strong> patient’snutritional <strong>and</strong> immune status, <strong>the</strong> number <strong>of</strong> reinfections,<strong>and</strong> <strong>the</strong> presence or absence <strong>of</strong> secondarybacterial infection. It is usually bilateral. The course<strong>of</strong> trachoma can be divided into four stages (<strong>the</strong> Mc-Callan classification) [68]. During <strong>the</strong> active stages <strong>of</strong>infection inclusions can be found in Giemsa-stainedcells scraped from <strong>the</strong> surface <strong>of</strong> infected epi<strong>the</strong>lium.More easy to recognise is a positive direct immun<strong>of</strong>luorescencefor Chlamydia trachomatis on <strong>the</strong> scraping,it can be very helpful in making <strong>the</strong> correct diagnosis[62, 90]. The diagnosis can also be made by isolation <strong>of</strong>


288 M.R. Canninga-Van Dijk10<strong>the</strong> causative agent in cell cultures <strong>and</strong> <strong>the</strong> detection <strong>of</strong>chlamydial antibody in blood or tear fluid. At Stage I<strong>the</strong>re is epi<strong>the</strong>lial infection by Chlamydia trachomatis.It is characterised clinically by <strong>the</strong> formation <strong>of</strong> conjunctivalfollicles <strong>and</strong> diffuse punctate inflammation<strong>of</strong> <strong>the</strong> cornea. The histology is indistinguishable fromthat <strong>of</strong> follicular conjunctivitis caused by o<strong>the</strong>r agents.Lymphocytes <strong>and</strong> plasma cells infiltrate <strong>the</strong> subepi<strong>the</strong>lialtissue; polymorphonuclear leukocytes infiltrate <strong>the</strong>corneal <strong>and</strong> conjunctival epi<strong>the</strong>lium. At Stage II, <strong>the</strong>inflammatory reaction occupies <strong>the</strong> stroma, with <strong>the</strong>fur<strong>the</strong>r formation <strong>of</strong> follicles. Large macrophages withphagocytised debris (Leber cells) are seen in <strong>the</strong> conjunctiva,accompanying <strong>the</strong> epi<strong>the</strong>lial hyperplasia withround cell infiltration <strong>and</strong> subepi<strong>the</strong>lial oedema [125].At Stage III <strong>the</strong> follicles disappear <strong>and</strong> cicatrisationoccurs. The fibrosis causes inversion <strong>of</strong> <strong>the</strong> upper lid(cicatricial entropion), misdirected lashes (trichiasis)<strong>and</strong> decreased tear formation. On histologic examination,scattered lymphocytes <strong>and</strong> plasma cells can stillbe seen along with subepi<strong>the</strong>lial scar tissue. At StageIV, <strong>the</strong>re is spontaneous arrest <strong>of</strong> <strong>the</strong> disease, which isno longer contagious. The residual entropion <strong>and</strong> trichiasislead to continuing corneal damage. Denuding<strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium leaves <strong>the</strong> cornea vulnerable to infection<strong>and</strong> fur<strong>the</strong>r opacification as a result <strong>of</strong> scarring.10.2.4.5.2 Inclusion ConjunctivitisThe conjunctival involvement <strong>of</strong> <strong>the</strong> sexually transmittedchlamydial infection is mild <strong>and</strong> can even be asymptomatic.In adults, it presents as a subacute follicularconjunctivitis. It is accompanied by a chronic urethritisin <strong>the</strong> male <strong>and</strong> a symptomless cervicitis in <strong>the</strong> female.In newborns, it occurs with an acute mucopurulentdischarge, 5 to 10 days after birth. It is accompaniedby infection <strong>of</strong> <strong>the</strong> maternal vagina by <strong>the</strong> same agent.Because <strong>the</strong> extranodal lymphoid tissues are not fullydeveloped, <strong>the</strong> conjunctivitis is more papillary than follicular.Like trachoma it can be diagnosed by Giemsastaining on conjunctival scrapings.10.2.5 Dermatologic<strong>and</strong> Systemic Diseases10.2.5.1 Keratoconjunctivitis SiccaIn this condition <strong>the</strong> cornea <strong>and</strong> conjunctiva are dry,causing a painful <strong>and</strong> gritty sensation. Keratoconjunctivitissicca was described first by Sjögren in 1933 [106,107]. It is <strong>the</strong>refore best known as one <strong>of</strong> <strong>the</strong> symptoms<strong>of</strong> Sjögren’s syndrome. However, it can also be seen ino<strong>the</strong>r auto-immune diseases like scleroderma or rheumatoidarthritis. Moreover, keratoconjunctivitis sicca is<strong>the</strong> most frequent cause <strong>of</strong> eye involvement in graft-versus-hostdisease [20, 50, 64]. Histologically, <strong>the</strong>re is atrophy<strong>of</strong> <strong>the</strong> lacrimal acinar parenchyma, accompanied byfibrosis <strong>and</strong> fatty infiltration, but with preservation <strong>of</strong><strong>the</strong> lobular architecture. There is a focal or diffuse presence<strong>of</strong> lymphocytes <strong>and</strong> plasma cells. Sometimes lymphoepi<strong>the</strong>liallesions can be seen (See Chap. 5 for moredetail.).10.2.5.2 Dermatologic DiseasesMany skin diseases can involve <strong>the</strong> conjunctiva. Mostfrequently seen are bullous diseases like pemphigus [72],bullous pemphigoid, Stevens-Johnson syndrome [43],paraneoplastic pemphigus [58, 74] <strong>and</strong> less commonly,dermatitis herpetiformis [43] <strong>and</strong> linear IgA disease [4].O<strong>the</strong>r dermatologic diseases with conjunctival involvementare lupus ery<strong>the</strong>matosus [40, 115, 122], familialchronic benign pemphigus (Hailey-Hailey disease) [80]<strong>and</strong> lichen planus [78, 116].10.2.5.3 Metabolic DiseasesA conjunctival biopsy can be <strong>of</strong> diagnostic value in metabolicdiseases with specific ultrastructural features,like galactosialidosis <strong>and</strong> different types <strong>of</strong> mucopolysaccharidoses[14, 121].10.2.6 Tumours<strong>and</strong> Tumour-Like Conditions10.2.6.1 Epi<strong>the</strong>lialEpi<strong>the</strong>lial tumours <strong>of</strong> <strong>the</strong> conjunctiva can be dividedinto tumours <strong>of</strong> <strong>the</strong> surface epi<strong>the</strong>lium (papilloma, intraepi<strong>the</strong>lialneoplasia <strong>and</strong> squamous cell carcinoma)<strong>and</strong> adnexal tumours. Since <strong>the</strong> caruncle contains accessorylacrimal gl<strong>and</strong>s, sweat gl<strong>and</strong>s, hair follicles <strong>and</strong>sebaceous gl<strong>and</strong>s, adnexal tumours <strong>of</strong> different kindscan be found.10.2.6.1.1 PapillomaICD-O:8560/0The commonest epi<strong>the</strong>lial tumours <strong>of</strong> <strong>the</strong> conjunctivaare papillomas, usually presenting as a red, papillomatousmass. These benign tumours histologicallyconsist <strong>of</strong> a fibrovascular core, lined with conjunctivalepi<strong>the</strong>lium, eventually with squamous metaplasia(Figs. 10.6, 10.7). The sessile variant usually shows only


Eye <strong>and</strong> Ocular Adnexa Chapter 10 289Fig. 10.6. Conjunctival papilloma: papillomatous mass consisting<strong>of</strong> a fibrovascular core covered with squamous metaplastic epi<strong>the</strong>liumFig. 10.8. Conjunctival intraepi<strong>the</strong>lial neoplasia: <strong>the</strong> epi<strong>the</strong>liumconsists <strong>of</strong> atypical cells without orderly differentiation<strong>of</strong> dysplasia, <strong>the</strong> epi<strong>the</strong>lial cells show atypia (Fig. 10.8). Inactinic keratosis <strong>the</strong> epi<strong>the</strong>lium is thin <strong>and</strong> <strong>the</strong> stromashows damage <strong>of</strong> elastic tissue. In <strong>the</strong> Bowenoid type mitoticfigures can be seen in <strong>the</strong> upper epi<strong>the</strong>lial layers <strong>and</strong>HPV-related epi<strong>the</strong>lial changes like multinucleated cells<strong>and</strong> koilocytes can be found [47, 71, 82].10.2.6.1.3 Malignant Tumours<strong>of</strong> <strong>the</strong> Surface Epi<strong>the</strong>liumFig. 10.7. Conjunctival papilloma: detail <strong>of</strong> <strong>the</strong> squamous metaplasticepi<strong>the</strong>liumSquamous cell carcinomas are rare <strong>and</strong> usually well differentiated.After adequate <strong>the</strong>rapy total cure is <strong>of</strong>tenachieved [13, 120]. More aggressive variants <strong>of</strong> this carcinoma,like <strong>the</strong> acantholytic or adenoid type [70] <strong>and</strong> <strong>the</strong>spindle cell type are even more rare. Mucoepidermoidcarcinoma <strong>of</strong> <strong>the</strong> conjunctiva has also been reported [34,101] (See Chaps. 1 <strong>and</strong> 5 for more details.).metaplastic epi<strong>the</strong>lium, without goblet cells. There is astrong association between HPV <strong>and</strong> conjunctival papillomas.HPV type 6/11 is <strong>the</strong> most common HPV typein conjunctival papilloma. The sensitivity <strong>of</strong> koilocytosisas an indicator <strong>of</strong> HPV in conjunctival papillomais low [105]. Papillomas are more extensively discussedin Chap. 1.10.2.6.1.2 ConjunctivalIntraepi<strong>the</strong>lial NeoplasiaIntraepi<strong>the</strong>lial neoplasia <strong>of</strong> <strong>the</strong> conjunctiva can be causedby sun-damage (actinic keratosis) or by HPV-induced premalignanttransformation (Bowenoid type). In both types10.2.6.1.4 OncocytomaICD-O:8290/0An oncocytoma (oxyphil cell adenoma, oxyphillicgranular cell adenoma) is a benign tumour that canoccur in <strong>the</strong> conjunctiva, <strong>the</strong> lacrimal gl<strong>and</strong> <strong>and</strong> <strong>the</strong>lacrimal sac [7, 111]. It presents as an asymptomatic,slowly progressive swelling <strong>of</strong> <strong>the</strong> caruncle in olderpersons. The lesion consists <strong>of</strong> large epi<strong>the</strong>lial cellswith eosinophilic, granular cytoplasm, due to a largenumber <strong>of</strong> mitochondria. The cells can be arrangedin cords, sheets or nests. Ductal <strong>and</strong> cystic gl<strong>and</strong>ularstructures can be found. The malignant variant <strong>of</strong> thistumour, <strong>the</strong> oncocytic adenocarcinoma <strong>of</strong> <strong>the</strong> lacrimalsystem is very rare.


290 M.R. Canninga-Van DijkFig. 10.9. Intraepi<strong>the</strong>lial component <strong>of</strong> a sebaceous adenocarcinoma:intraepi<strong>the</strong>lial pagetoid spread <strong>of</strong> tumour cells may be misinterpretedas dysplasia; positive staining for EMA can be veryhelpful (EMA immunostaining)Fig. 10.11. Sebaceous adenocarcinoma: detail <strong>of</strong> <strong>the</strong> clear cells,showing sebaceous differentiation10like EMA <strong>and</strong> CAM 5.2 can help in differentiating thisaggressive tumour from a squamous cell carcinoma(Figs. 10.09–10.11). Treatment <strong>of</strong> choice is wide excision,which can cure patients at an early stage <strong>of</strong> <strong>the</strong>lesion. However, <strong>the</strong> mortality rate from metastases is25%, <strong>and</strong> even higher in a poorly differentiated tumourwith angioinvasive growth.10.2.6.2 Melanocytic10.2.6.2.1 NaevusFig. 10.10. Sebaceous adenocarcinoma: basaloid epi<strong>the</strong>lial nestswith a clear cell component10.2.6.1.5 Sebaceous AdenocarcinomaICD-O:8410/3This lesion is important because it can be a pitfall forboth <strong>the</strong> clinician <strong>and</strong> <strong>the</strong> pathologist. The tumourpresents as a solitary nodule, that can clinically be misdiagnosedas a basal cell carcinoma or even as a chalazionor blepharoconjunctivitis [1, 36, 81]. Histologically,<strong>the</strong> tumour is composed <strong>of</strong> epi<strong>the</strong>lial nests withvarying sebaceous differentiation. The well-differentiatedsebaceous carcinomas are not very hard to recognise,but <strong>the</strong> poorly differentiated ones can be easilymissed. The intraepi<strong>the</strong>lial pagetoid spread <strong>of</strong> tumourcells (which is frequently present) may be misinterpretedas dysplasia. Immunohistochemical stainingsICD-O:8720/0The most common melanocytic lesion <strong>of</strong> <strong>the</strong> conjunctivais <strong>the</strong> compound naevus. O<strong>the</strong>r types <strong>of</strong> naevi thatcan be found in <strong>the</strong> conjunctiva are intraepi<strong>the</strong>lial,subepi<strong>the</strong>lial, Spitz <strong>and</strong> blue naevi. Their histology issimilar to melanocytic skin lesions. The naevus mostlyarises in <strong>the</strong> first or second decade as a nodule in <strong>the</strong>bulbar conjunctiva. A b<strong>and</strong> <strong>of</strong> melanocytes in <strong>the</strong> basallayer <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium represents <strong>the</strong> intraepi<strong>the</strong>lialcomponent. These melanocytes can be melanin-containing,but can also present as clear cells. Melanocytescan also be found in <strong>the</strong> epi<strong>the</strong>lium <strong>of</strong> <strong>the</strong> inclusioncysts, which are almost invariably present. These large,mucin-containing cysts are formed by incarceratedepi<strong>the</strong>lial nests <strong>and</strong> can give an erroneous clinical impression<strong>of</strong> growth. The stromal component is formedby nests <strong>of</strong> mature cells with maturation to smaller cellsin <strong>the</strong> deeper parts <strong>of</strong> <strong>the</strong> lesion (Fig. 10.12). Especiallyat a young age, a considerable variation in cell size canbe seen; <strong>the</strong>se active lesions are easily overdiagnosed asmalignant melanomas.


Eye <strong>and</strong> Ocular Adnexa Chapter 10 291Fig. 10.12. Compound conjunctival naevus: <strong>the</strong> intraepi<strong>the</strong>lialcomponent is represented by a b<strong>and</strong> <strong>of</strong> clear melanocytes in <strong>the</strong>basal layer <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium. A small inclusion cyst is present, surroundedby unsuspicious naevoid cells with maturation to smallercells in <strong>the</strong> deeper part <strong>of</strong> <strong>the</strong> lesionFig. 10.14. Malignant melanoma arising from primary acquiredmelanosis with atypia: <strong>the</strong> conjunctival epi<strong>the</strong>lium is ulcerated<strong>and</strong> <strong>the</strong> atypical cells spread into <strong>the</strong> underlying stromaspontaneously, can remain stationary or may progress tomalignant melanoma. Histologically, <strong>the</strong>re is an increasein atypical melanocytes in <strong>the</strong> conjunctival epi<strong>the</strong>lium(Fig. 10.13). The atypia can be graded mild to severe. Toexclude invasive growth, use <strong>of</strong> <strong>the</strong> immunohistochemicalmarker CD68 can be helpful in identifying melanin-containingcells in <strong>the</strong> stroma as macrophages.10.2.6.2.3 Malignant MelanomaFig. 10.13. Primary acquired melanosis with atypia: an almostcontinuous proliferation <strong>of</strong> atypical melanocytes is present in <strong>the</strong>conjunctival epi<strong>the</strong>lium10.2.6.2.2 Primary Acquired MelanosisICD-O:8741/2Primary acquired melanosis (PAM) arises in middle-agedor elderly patients as a stippled, yellow-brown, flat pigmentation<strong>of</strong> <strong>the</strong> conjunctiva. Two subgroups <strong>of</strong> PAM can berecognised: PAM without atypia (benign acquired melanosis)<strong>and</strong> PAM with atypia. In benign acquired melanosis,<strong>the</strong>re is hyperpigmentation <strong>of</strong> <strong>the</strong> basal layer, but <strong>the</strong>re isonly a mild increase in melanocytes. The melanocytes canbe large, but show little or no cytologic atypia. Although <strong>the</strong>evolution <strong>of</strong> PAM is unpredictable, lesions without atypiahave a good prognosis. PAM with atypia can disappearICD-O:8720/3The majority <strong>of</strong> conjunctival melanomas arise withinprimary acquired melanosis with atypia. Development<strong>of</strong> a melanoma in a pre-existing naevus or de novo ispossible, but uncommon [44, 61]. In malignant melanomaclusters <strong>of</strong> atypical melanocytes are present in <strong>the</strong>stroma (Figs. 10.14, 10.15). The melanocytes are mostfrequently epi<strong>the</strong>lioid, but can also be spindle-shapedor bizarre. The intraepi<strong>the</strong>lial component shows large,atypical melanocytes, <strong>of</strong>ten without ascending cells.This differs from skin melanocytic lesions, where ascendingmelanocytes can be very helpful in diagnosinga malignant melanoma. These cytological characteristicsdo not seem to influence prognosis. Depth <strong>of</strong> <strong>the</strong>tumour, however, does have prognostic value: thicknessless than 1.5 mm means a low risk <strong>of</strong> metastatic disease.10.2.6.3 O<strong>the</strong>r NeoplasmsLymphomas may involve <strong>the</strong> conjunctiva. Also, tumoursfrom adjacent locations may extend into this site. Theyare discussed under <strong>the</strong>ir appropriate headings.


292 M.R. Canninga-Van DijkFig. 10.15. Malignant melanoma: detail <strong>of</strong> <strong>the</strong> nested, atypicalmelanocytes without maturation in <strong>the</strong> stromaFig. 10.16. Mucopolysaccharidosis: this cornea showed no abnormalitieson H&E staining, but colloidal iron showed deposits <strong>of</strong>mucopolysaccharides very clearly1010.3 CorneaInflammation <strong>and</strong> ulceration <strong>of</strong> <strong>the</strong> cornea can becaused by trauma, surgery to <strong>the</strong> eye, infectious diseases<strong>and</strong> systemic diseases. The trauma can be mechanical,chemical or caused by heat or irradiation. The increasingincidence <strong>of</strong> cataract extraction has led to an increasein cases with corneal damage during <strong>the</strong> surgical procedure.Infectious keratitis can be viral, bacterial, fungalor parasitic. In chronic granulomatous keratitis, causeslike leprosy, syphilis <strong>and</strong> tuberculosis should be considered.Especially in immunocompromised patients infectiouscauses should be excluded by additional stainslike PAS, Grocott <strong>and</strong> Gram. In corneal ulcers withouta clear aetiology, artificial keratitis has to be considered.Factitious injury is rare <strong>and</strong> can be ei<strong>the</strong>r <strong>the</strong> result <strong>of</strong>mechanical trauma or <strong>the</strong> abuse <strong>of</strong> toxic eye drops [26].Histology <strong>of</strong> factitious lesions is non-specific. An example<strong>of</strong> a systemic disease affecting <strong>the</strong> cornea is mucopolysaccharidosis(Fig. 10.16).10.3.1 Keratitis<strong>and</strong> Corneal Ulcersor punctate spots (punctate keratopathy). If <strong>the</strong> diseaserecurs, <strong>the</strong> virus may infect stromal keratocytes, causingchronic destruction with ulceration. Histologically,an early herpetic ulcer shows multinucleated epi<strong>the</strong>lialcells with intranuclear viral inclusions. DNA in situhybridisation, immunohistochemistry <strong>and</strong> PCR aremodern techniques replacing <strong>the</strong> transmission electronmicroscopy that was used to demonstrate <strong>the</strong>herpes virus particles in earlier days [49]. In end-stagedisease, epi<strong>the</strong>lial changes are no longer present; <strong>the</strong>reis just fibrosis with scarring. Often, Bowman’s layer isfocally replaced by fibrosis.10.3.1.2 Corneal UlcerationDue to Systemic DiseaseSystemic vasculitides like systemic lupus ery<strong>the</strong>matosus,polyarteritis nodosa <strong>and</strong> Wegener’s granulomatosiscan cause peripheral corneal ulceration due to vascularocclusion by immune complex deposition in <strong>the</strong>limbal vessels [73, 85]. In rheumatoid arthritis cornealulceration can occur due to <strong>the</strong> release <strong>of</strong> collagenases[92].10.3.1.1 Herpes Simplex KeratitisType 1 herpes simplex virus is <strong>the</strong> most common cause<strong>of</strong> corneal disease. After primary infection <strong>of</strong> <strong>the</strong> lip,<strong>the</strong> virus remains latent in <strong>the</strong> sensory trigeminal ganglion.Transneural migration <strong>and</strong> proliferation <strong>of</strong> <strong>the</strong>virus is triggered by stress, sunlight or cold. The virusparasitises <strong>the</strong> epi<strong>the</strong>lium, leading to superficial epi<strong>the</strong>lialloss in a branching pattern (dendritic ulceration)10.3.2 KeratoconusThis condition presents at puberty <strong>and</strong> has been foundin association with systemic disorders like Marfan’s syndrome,Down’s syndrome [52, 88], neur<strong>of</strong>ibromatosis,Ehlers-Danlos syndrome [56, 94, 129] <strong>and</strong> atopic dermatitis[93, 109, 119]. It can also be seen in combinationwith ocular disorders like aniridia, cataract <strong>and</strong> retinitispigmentosa [11, 33, 53]. A progressive, non-inflamma-


Eye <strong>and</strong> Ocular Adnexa Chapter 10 293tory, bilateral thinning <strong>of</strong> <strong>the</strong> central corneal stromaleads to severe astigmatism. Aetiology <strong>and</strong> pathogenesisare unclear. At histological examination <strong>the</strong> epi<strong>the</strong>liumcan be ei<strong>the</strong>r atrophic or hyperplastic. The most strikingfinding is interruption <strong>of</strong> Bowman’s membrane, withdowngrowths <strong>of</strong> epi<strong>the</strong>lium or upgrowths <strong>of</strong> cornealstroma in <strong>the</strong> breaking spot. The breaks may be narrow<strong>and</strong> <strong>the</strong> pathology is <strong>of</strong>ten restricted to a narrow 1–2 mmzone, sometimes serial sections are required to find <strong>the</strong>lesion. At <strong>the</strong> edge <strong>of</strong> <strong>the</strong> conus, iron can be found in <strong>the</strong>epi<strong>the</strong>lium (Fleischer’s ring). The axial stroma showsmucoid degeneration, <strong>the</strong> peripheral stroma is <strong>of</strong> normalappearance. In severe cases, rupture <strong>of</strong> Descemet’smembrane <strong>and</strong> <strong>the</strong> endo<strong>the</strong>lium can occur, resulting inan inflow <strong>of</strong> water <strong>and</strong> <strong>the</strong> appearance <strong>of</strong> cystic spaces.Keratoconus can be treated by surgery with use <strong>of</strong> stromalcornea grafts.10.3.3 Hereditary Corneal DystrophiesCorneal dystrophies are inherited, bilateral disordersthat can be divided into epi<strong>the</strong>lial, stromal <strong>and</strong> endo<strong>the</strong>lialabnormalities. Routine stains for suspected cornealdystrophy must include PAS, Masson, Alcian blue,Congo red <strong>and</strong> Trichrome stains. In end-stage dystrophiesa keratopathy can develop, in which all layers <strong>of</strong><strong>the</strong> cornea are involved. Many patients with cornealdystrophies have a point mutation in a gene on chromosome5q31 [54, 77, 113].10.3.3.1 Epi<strong>the</strong>lial DystrophiesEpi<strong>the</strong>lial corneal dystrophies are Cogan’s microcysticdystrophy, Meesman’s dystrophy <strong>and</strong> Reis-Buckler’sring dystrophy [123]. The epi<strong>the</strong>lial dystrophies presentwith photophobia <strong>and</strong>/or foreign body sensation.The most common form <strong>of</strong> epi<strong>the</strong>lial dystrophy isCogan’s microcystic dystrophy, affecting females <strong>of</strong>middle age. A thickened <strong>and</strong> folded basement membranewith epi<strong>the</strong>lial cysts containing necrotic debrisis characteristic. Bowman’s membrane is not involved.In Meesman’s dystrophy small layers <strong>of</strong> <strong>the</strong> basementmembrane can be found between <strong>the</strong> epi<strong>the</strong>lial cells.Reis-Buckler ring dystrophy is a bilateral, autosomaldominant dystrophy, not only affecting <strong>the</strong> epi<strong>the</strong>lium,but also <strong>the</strong> anterior corneal stroma. The epi<strong>the</strong>lium isoedematous <strong>and</strong> atrophic, Bowman’s membrane is interrupted<strong>and</strong> <strong>the</strong> anterior stroma contains abnormalfibrous tissue. Histology is not specific <strong>and</strong> transmissionelectron microscopy, which will show electrondenserods in <strong>the</strong> superficial stroma, is necessary toconfirm <strong>the</strong> diagnosis [87].10.3.3.2 Stromal Dystrophies10.3.3.2.1 Granular DystrophyGranular dystrophy is an autosomal dominant disorder,presenting in early childhood with discrete, opaquegranules in <strong>the</strong> o<strong>the</strong>rwise transparent anterior cornealstroma. Histologically, non-birefringent hyaline bodiesare present in <strong>the</strong> stroma. The deposits are strongly positivewith Masson stain [67].10.3.3.2.2 Lattice DystrophyLattice dystrophy is an autosomal dominant disorder, clinicallypresenting in early childhood (type I) [66] or in <strong>the</strong>2nd decade (type II) [95] with linear opacities. Histologically,eosinophilic deposits are found in <strong>the</strong> corneal stroma.They consist <strong>of</strong> amyloid <strong>and</strong> are strongly Congo red-positive[67, 112]. The disease is treated by keratoplasty.The presence <strong>of</strong> amyloid in <strong>the</strong> corneal stroma canalso be seen in chronic inflammatory conditions, <strong>the</strong>sesecondary amyloid deposits should not be confused withlattice dystrophy.10.3.3.2.3 Avellino DystrophyA combined granular-lattice dystrophy with both hyaline<strong>and</strong> amyloid deposits in <strong>the</strong> corneal stroma was firstdescribed in <strong>the</strong> Italian village <strong>of</strong> Avellino [29].10.3.3.2.4 Macular Corneal DystrophyMacular dystrophy presents in childhood as a bilateralprocess with irregular opacities between cloudy cornealstroma. It is a disease disabling vision, inherited as anautosomal recessive trait. The disease is considered to bea localised metabolic disorder with production <strong>of</strong> excessiveamounts <strong>of</strong> acid mucopolysaccharide by fibroblasts[24, 67]. The disease is not associated with systemic mucopolysaccharidoses.10.3.3.3 Endo<strong>the</strong>lial Dystrophies10.3.3.3.1 Fuchs DystrophyThis disorder affects elderly patients <strong>and</strong> is quite commonin routine histopathology. It presents clinicallywith bilateral diffuse cloudy <strong>and</strong> oedematous stroma.Histologically, <strong>the</strong> corneal epi<strong>the</strong>lium is oedematous,


294 M.R. Canninga-Van Dijk10Descemet’s membrane is thickened <strong>and</strong> <strong>the</strong>re is reduction<strong>of</strong> <strong>the</strong> endo<strong>the</strong>lial cell population. A PAS stainsometimes shows lamination <strong>of</strong> Descemet’s membrane.On <strong>the</strong> posterior surface <strong>of</strong> Descemet’s membrane, nodularexcrescences can be seen.10.3.4 Failed Previous GraftsMany corneal diseases can be treated by keratoplasty.Complications are rejection, formation <strong>of</strong> a retrocornealfibrous membrane <strong>and</strong> recurrence <strong>of</strong> <strong>the</strong> original disease.Rejection <strong>of</strong> <strong>the</strong> graft can occur immediately after<strong>the</strong> operation or many years later. Histologically, <strong>the</strong>re isvascularisation <strong>of</strong> <strong>the</strong> corneal stroma, accompanied by alymphocytic infiltrate.Formation <strong>of</strong> retrocorneal fibrous membranes occursfollowing fibroblastic metaplasia <strong>of</strong> keratocytes at <strong>the</strong>posterior edges <strong>of</strong> <strong>the</strong> host-graft junction. This complicationis diminishing with advances in microsurgery.Over a longer period <strong>of</strong> time, recurrence <strong>of</strong> <strong>the</strong> originaldisease is common in patients with corneal dystrophies.10.4 Intraocular Tissues<strong>Pathology</strong> <strong>of</strong> <strong>the</strong> intraocular tissues can be divided intodevelopmental anomalies, inflammatory processes,trauma, degeneration <strong>and</strong> tumours.10.4.1 Developmental Anomalies10.4.1.1 Congenital GlaucomaGlaucoma due to congenital malformation is rare. Associationswith systemic disorders like neur<strong>of</strong>ibromatosis[8, 84, 130] <strong>and</strong> Sturge-Weber syndrome [18, 19] havebeen described. A malformation <strong>of</strong> <strong>the</strong> trabecular meshwork(goniodysgenesis) or persistence <strong>of</strong> embryonic tissuein <strong>the</strong> chamber angle causes an outflow obstruction.The corneoscleral envelope <strong>of</strong> <strong>the</strong> infant is distensible,so that a raised intraocular pressure can produce an enlargement<strong>of</strong> <strong>the</strong> globe (buphthalmos). A hypercellulartrabecular meshwork with hyaloid degeneration is <strong>the</strong>best visible histopathological finding in those specimens[133].10.4.1.2 Retinopathy <strong>of</strong> PrematurityIn premature children requiring artificial breathingwith high oxygen pressures to survive, disordered neovascularisationat <strong>the</strong> periphery <strong>of</strong> <strong>the</strong> retina can occur[91]. A white retrolental fibrous membrane can form in<strong>the</strong> most extreme cases, causing bilateral blindness ( retrolentalfibroplasia). At <strong>the</strong> end stage <strong>of</strong> <strong>the</strong> disease neovascularglaucoma leads to enucleation. The distortedretina is macroscopically visible, most frequently forminga straight b<strong>and</strong> behind <strong>the</strong> lens. Histology showsretinal gliosis <strong>and</strong> optic atrophy [21].10.4.1.3 Persistent PrimaryHyperplastic VitreousIn <strong>the</strong> embryo, <strong>the</strong> lens is supported posteriorly by amass <strong>of</strong> vascular tissue, <strong>the</strong> primary vitreous. If this primaryvitreous fails to involute before birth, embryonicfibrovascular tissue persists in <strong>the</strong> anterior or posteriorpart <strong>of</strong> <strong>the</strong> vitreous <strong>and</strong> becomes hyperplastic. In persistentanterior primary hyperplastic vitreous, a retrolentalfibrovascular mass can penetrate <strong>the</strong> posterior lens capsule<strong>and</strong> lens cortex, causing an autoimmune inflammatoryresponse. In persistent posterior hyperplasticprimary vitreous, <strong>the</strong> fibrovascular mass damages <strong>the</strong>optic disc.10.4.1.4 Retinal DysplasiaFailure <strong>of</strong> organisation <strong>of</strong> <strong>the</strong> layers <strong>of</strong> <strong>the</strong> retina leadsto retinal dysplasia, in which nests <strong>of</strong> neuroblastic cellsform rosettes within <strong>the</strong> retina. Clustering <strong>of</strong> rosettesleads to thickening <strong>of</strong> <strong>the</strong> retina, which may become detached.Retinal dysplasia is a common feature <strong>of</strong> trisomy13, trisomy 18 <strong>and</strong> o<strong>the</strong>r chromosomal disorders. As anisolated entity, retinal dysplasia is very rare [65].10.4.1.5 AniridiaAniridia is a rare bilateral disease. It occurs as a conditionthat may be inherited as an autosomal dominantdisorder or as part <strong>of</strong> several systemic syndromes. Insome cases <strong>of</strong> aniridia, deletion <strong>of</strong> <strong>the</strong> short arm <strong>of</strong>chromosome 11 occurs. Because this locus lies closeto <strong>the</strong> gene for nephroblastoma (Wilms’ tumour), <strong>the</strong>recognition <strong>of</strong> a child with sporadic aniridia shouldalert physicians to <strong>the</strong> increased risk <strong>of</strong> development <strong>of</strong>Wilms’ tumour [10, 23]. Histology <strong>of</strong> enucleated eyesor trabeculectomy specimens shows a rudimentaryiris, consisting <strong>of</strong> hypercellular stroma, <strong>of</strong>ten with anabnormal proliferation <strong>of</strong> pigment epi<strong>the</strong>lium.10.4.1.6 Congenital Rubella SyndromeMaternal rubella infection during <strong>the</strong> first trimester <strong>of</strong>pregnancy can affect <strong>the</strong> development <strong>and</strong> function <strong>of</strong><strong>the</strong> entire eye. It can manifest as a congenital cataract,


Eye <strong>and</strong> Ocular Adnexa Chapter 10 295retinitis can also be visualised with DNA in situ hybridisation.10.4.2.2 Chronic Non-GranulomatousInflammationChronic non-granulomatous inflammation <strong>of</strong> <strong>the</strong> uvealtract is a poorly understood condition. It can be dividedinto anterior uveitis, with risk <strong>of</strong> secondary angle closureglaucoma, <strong>and</strong> posterior uveitis with risk <strong>of</strong> degeneration<strong>of</strong> retinal pigment epi<strong>the</strong>lium. The histology isnon-specific, with only a few lymphocytes in <strong>the</strong> uvealtissues.Fig. 10.17. Cytomegaloviral retinitis: large eosinophilic inclusionsare present in <strong>the</strong> infected endo<strong>the</strong>lial cells. In this biopsy<strong>the</strong> retina showed almost no signs <strong>of</strong> necrosis, which is uncommonin cytomegalovirus retinitisdisciform keratitis, retinopathy, microphthalmus oropen-angle glaucoma [3, 51]. In societies with immunisation<strong>the</strong> condition is rare. The histologic findings in<strong>the</strong> lens are characteristic, but it should be mentionedthat <strong>the</strong> features may not yet be apparent in foetal eyesafter early elective termination <strong>of</strong> pregnancy [38]. Thecentral nucleus <strong>of</strong> <strong>the</strong> lens, which is normally free <strong>of</strong>cells, shows pyknotic nuclei. There is an abrupt transitionfrom <strong>the</strong> central nucleus into <strong>the</strong> normal peripheralcortex <strong>of</strong> <strong>the</strong> lens [128].10.4.2 Inflammatory Processes10.4.2.1 Acute InflammationAcute endophthalmitis or panophthalmitis can occuras a postoperative complication or following a trauma[12, 57]. Endocarditis or injection <strong>of</strong> contaminatedmaterial in drug addicts can be <strong>the</strong> cause <strong>of</strong> metastaticbacterial infection, especially in immunocompromisedpatients [89]. Not only bacterial, but also fungal <strong>and</strong>viral infections can cause acute endophthalmitis. Histologyshows an extensive infiltrate <strong>of</strong> leukocytes withdestruction <strong>of</strong> intraocular tissues. The pathogen cansometimes be found in Gram, PAS or Silver stains.Acute necrotising retinitis <strong>and</strong> low-grade uveitiscan be seen in cytomegaloviral <strong>and</strong> herpes infection(Fig. 10.17). The characteristic eosinophilic inclusions<strong>of</strong> <strong>the</strong> cytomegalovirus can be found in <strong>the</strong> cytoplasm<strong>and</strong> nuclei <strong>of</strong> infected cells. Immunohistochemicalstaining with anti-cytomegalovirus can be helpful inidentifying <strong>the</strong> virus. The inclusions <strong>of</strong> herpes simplex10.4.2.3 Granulomatous InflammationThe specific granulomatous inflammations <strong>of</strong> <strong>the</strong> intra-oculartissues can be divided into infectious <strong>and</strong>autoimmune causes. The autoimmune diseases are sarcoidosis,sympa<strong>the</strong>tic ophthalmitis <strong>and</strong> lens-induceduveitis. While many <strong>of</strong> <strong>the</strong>se diseases may be appropriatelytreated with immunosuppressive medication, <strong>the</strong>management <strong>of</strong> infectious uveitis is antimicrobial <strong>the</strong>rapy.Inappropriate immunosuppressive <strong>the</strong>rapy may bedisastrous for patients with an infection. Chorioretinalbiopsy may provide useful information for determining<strong>the</strong> diagnosis <strong>and</strong> guiding <strong>the</strong> subsequent management<strong>of</strong> patients with progressive chorioretinal lesions <strong>of</strong> unknownaetiology [69].10.4.2.3.1 InfectiousThe most important causes <strong>of</strong> infectious granulomatousinflammatory diseases <strong>of</strong> <strong>the</strong> intraocular tissues are tuberculosis<strong>and</strong> toxoplasmosis.Tuberculosis is rare <strong>and</strong> shows caseating granulomas,in which tubercle bacilli can be found in a Ziehl-Neelsenstaining.Toxoplasmic retinochoroiditis in neonates infectedin utero with Toxoplasma gondii can show a wide variationin <strong>the</strong> pattern <strong>of</strong> tissue destruction. The disease canbe limited to a low grade uveitis <strong>and</strong> retinal lymphocyticperivasculitis. In more severely affected eyes focal, sectorialor total retinal destruction can be seen. Toxoplasmacysts can be found in <strong>the</strong> retina <strong>and</strong> optic nerve. In<strong>the</strong> most severely affected eyes <strong>the</strong> retina is necrotic <strong>and</strong>calcified [5].10.4.2.3.2 SarcoidosisIn patients with systemic sarcoidosis, ocular involvementcan occur. Most frequently affected are <strong>the</strong> retina, <strong>the</strong>


296 M.R. Canninga-Van Dijk10uveal tract <strong>and</strong> <strong>the</strong> optic nerve. Histology shows sharplydemarcated, non-caseating granulomas [59, 96].10.4.2.3.3 Sympa<strong>the</strong>tic OphthalmitisSympa<strong>the</strong>tic ophthalmitis is an uncommon, but fearedcomplication because <strong>of</strong> its potential to blind bo<strong>the</strong>yes. It can result not only from penetrating trauma orocular surgery, but also from non-penetrating ocularprocedures. Early enucleation <strong>of</strong> <strong>the</strong> traumatised eyereduces <strong>the</strong> risk <strong>of</strong> occurrence <strong>of</strong> sympa<strong>the</strong>tic ophthalmitisin <strong>the</strong> non-traumatised (“sympa<strong>the</strong>tic”) eye.The condition seems to be caused by a T-cell-mediatedautoimmune response <strong>and</strong> can be treated with immunosuppressive<strong>the</strong>rapy. Infectious causes must be ruledout before starting this <strong>the</strong>rapy. Histology <strong>of</strong> <strong>the</strong> enucleatedtraumatised eye shows a granulomatous uveitiswith a thickened choroid featuring non-caseatinggranulomas with a few plasma cells <strong>and</strong> eosinophils,very similar to sarcoidosis [16]. Fine melanin granulescan be seen in <strong>the</strong> cytoplasm <strong>of</strong> <strong>the</strong> histiocytes.The granulomatous inflammatory reaction can spreadaround small nerves.10.4.2.3.4 Lens-Induced UveitisLens-induced uveitis or phacoanaphylactic endophthalmitisis a chronic endophthalmitis with a zonal granulomatousinflammation surrounding a ruptured lens.Most cases occur after trauma, surgical or non-surgical.The condition may result in vision-threatening intraocularinflammation that is poorly responsive to medicalmanagement. Leaking <strong>of</strong> lens proteins through an intactlens capsule may result in a lympho-plasmacytic anterioruveitis [102, 127]. The inflammation can be confinedto <strong>the</strong> anterior aspect <strong>of</strong> <strong>the</strong> eye, but <strong>the</strong> choroid canalso be involved. Surgical removal <strong>of</strong> <strong>the</strong> lens materialis generally indicated shortly after <strong>the</strong> injury in an effortto save vision. Normally, small amounts <strong>of</strong> circulatinglens proteins maintain a normal T-cell tolerance for lensproteins. Lens-induced uveitis develops when a breakdownoccurs <strong>of</strong> this normal T-cell tolerance. Immunecomplexes play an important role in <strong>the</strong> tissue damageassociated with <strong>the</strong> ensuing inflammation.10.4.3 TraumaMechanical injury is <strong>the</strong> most frequent cause <strong>of</strong> trauma<strong>of</strong> <strong>the</strong> eye. It can be caused by many different forces, likebroken glass, airguns, knives <strong>and</strong> golf balls. Chemical,toxic <strong>and</strong> radiation damage is less <strong>of</strong>ten seen. Traumatisedeyes can be enucleated at three different momentsin <strong>the</strong> time following <strong>the</strong> injury.The first moment to make <strong>the</strong> decision for enucleation isimmediately or within a day or two following <strong>the</strong> injury.The globe is ruptured, massive intraocular bleeding ispresent <strong>and</strong> <strong>the</strong>re is evidence that repair will not restorevisual function. To lower <strong>the</strong> risk <strong>of</strong> sympa<strong>the</strong>tic ophthalmitis,<strong>the</strong> eye will be enucleated at an early phase.A wide range <strong>of</strong> foreign bodies can be found in <strong>the</strong>seeyes. The inflammation is usually mild or absent. Themost important mission for <strong>the</strong> pathologist is to confirm<strong>the</strong> irreparable damage. The lens is <strong>of</strong>ten absent or prolapsedthrough <strong>the</strong> corneal wound, a retinal tear shows<strong>the</strong> side <strong>of</strong> penetration, <strong>the</strong> vitreous is haemorrhagic,papilloedema is present <strong>and</strong> <strong>the</strong> retina can show exudativedetachment (Fig. 10.18).When attempts to repair <strong>the</strong> eye are made, mild uveitisdevelops. Within a period <strong>of</strong> 2–3 weeks <strong>the</strong> uveitisshould diminish. If not, many ophthalmologists willmake <strong>the</strong> decision to enucleate <strong>the</strong> eye to avoid <strong>the</strong> risk<strong>of</strong> sympa<strong>the</strong>tic ophthalmitis. In <strong>the</strong>se eyes, removedwithin a few weeks after <strong>the</strong> trauma, reparative changeslike fibrous ingrowth <strong>of</strong> <strong>the</strong> corneal wound can befound. The blood in <strong>the</strong> vitreous will show organisation.Traumatised eyes with residual vision <strong>and</strong> withoutinflammatory complications can become hypotonic <strong>and</strong>atrophic over a period <strong>of</strong> years. Secondary glaucoma c<strong>and</strong>evelop <strong>and</strong> <strong>the</strong> eyes are removed because <strong>of</strong> pain or forcosmetic reasons. Frequently, <strong>the</strong> secondary changes arevery complicated <strong>and</strong> <strong>the</strong> primary pathology is not visibleanymore. At macroscopic examination, <strong>the</strong> site <strong>of</strong><strong>the</strong> trauma can be identified by <strong>the</strong> presence <strong>of</strong> scars,suture tracks or just by episcleral thickening. Post-traumaticglaucoma is most <strong>of</strong>ten caused by secondary angleclosure. Fur<strong>the</strong>rmore, dislocation <strong>of</strong> <strong>the</strong> lens <strong>and</strong> lensinduceduveitis can be seen. The retina is usually partiallyor totally detached, <strong>and</strong> is thickened by reactivegliosis.10.4.4 Degeneration10.4.4.1 GlaucomaThe normal pressure (13–21 mmHg) in <strong>the</strong> corneoscleralenvelope is maintained by a balance between<strong>the</strong> aqueous inflow <strong>and</strong> <strong>the</strong> resistance in <strong>the</strong> outflowsystem. The fluid is pumped into <strong>the</strong> eye by <strong>the</strong> ciliaryepi<strong>the</strong>lium, passes through <strong>the</strong> pupil to <strong>the</strong> chamberangle <strong>and</strong> leaves <strong>the</strong> eye via <strong>the</strong> trabecular meshwork<strong>and</strong> <strong>the</strong> canal <strong>of</strong> Schlemm to <strong>the</strong> collector canals in<strong>the</strong> sclera, draining into <strong>the</strong> episcleral venous system.Impaired outflow can cause high intraocular pressures<strong>and</strong> when <strong>the</strong> intraocular pressure is high enough tocause damage to <strong>the</strong> intraocular tissues, <strong>the</strong> term glaucomais used. The primary cause <strong>of</strong> glaucoma can in


Eye <strong>and</strong> Ocular Adnexa Chapter 10 29710.4.4.1.2 Primary AngleClosure GlaucomaIn primary angle closure glaucoma, <strong>the</strong> aqueous outflowis obstructed by apposition <strong>of</strong> <strong>the</strong> iris to <strong>the</strong> innersurface <strong>of</strong> <strong>the</strong> cornea <strong>and</strong> <strong>the</strong> trabecular meshwork. Theacute form <strong>of</strong> <strong>the</strong> disease occurs unilaterally in middleaged<strong>and</strong> elderly patients <strong>and</strong> presents with a rapid <strong>and</strong>painful rise in intraocular pressure. Both in acute <strong>and</strong>chronic angle closure glaucoma, three ageing processesseem to cause <strong>the</strong> closure <strong>of</strong> <strong>the</strong> angle: shrinkage <strong>of</strong> <strong>the</strong>eye, reduction in depth <strong>of</strong> <strong>the</strong> anterior chamber <strong>and</strong> increasedsize <strong>of</strong> <strong>the</strong> lens.Fig. 10.18. Trauma caused by a wooden stick: this eye was enucleated1 week after <strong>the</strong> trauma <strong>and</strong> showed scleral perforation, retinaldetachment <strong>and</strong> haemorrhagic vitreousmost cases not be detected in <strong>the</strong> tissue without <strong>the</strong>complete clinical history. Depending upon <strong>the</strong> rapidity<strong>of</strong> <strong>the</strong> rise in pressure, glaucoma causes tissue damage.In a pressure rise to 80 mmHg within 1 or 2 days(acute glaucoma) severe corneal oedema, infarction <strong>of</strong><strong>the</strong> iris, necrosis <strong>of</strong> <strong>the</strong> lens <strong>and</strong> retinal oedema occur.When <strong>the</strong> pressure rises over a longer period <strong>of</strong> timemore chronic changes can be found. The cornea showsfibrovascular tissue ingrowth at <strong>the</strong> periphery. The angleis closed by a corneal endo<strong>the</strong>lial downgrowth <strong>and</strong><strong>the</strong> trabecular tissue is fused <strong>and</strong> hyalinised. The irisstroma <strong>and</strong> ciliary body become atrophic <strong>and</strong> fibrotic.The nucleus <strong>of</strong> <strong>the</strong> lens becomes sclerotic. The vitreousmay be unaffected, but in cases <strong>of</strong> retinal vasculardisease, <strong>the</strong> vitreous contains blood <strong>and</strong> macrophageswith fibrous str<strong>and</strong>s. Atrophy <strong>of</strong> <strong>the</strong> optic disc is visibleby cupping <strong>and</strong> shrinkage down to <strong>the</strong> lamina cribrosa,which becomes bowed posteriorly. The choroid <strong>and</strong>retinal pigment epi<strong>the</strong>lium are able to withst<strong>and</strong> highpressure <strong>and</strong> will only show atrophy <strong>and</strong> fibrosis inend-stage disease. Pathologic examination <strong>of</strong> enucleatedglaucomatous eyes is <strong>of</strong>ten complicated by previoussurgical procedures. Glaucoma can be divided int<strong>of</strong>our subgroups.10.4.4.1.1 Primary OpenAngle GlaucomaPrimary open angle glaucoma occurs predominantlyin <strong>the</strong> elderly <strong>and</strong> is caused by an acquired unilateral orbilateral disease <strong>of</strong> <strong>the</strong> trabecular meshwork, visible athistopathological examination by hyalinisation <strong>of</strong> <strong>the</strong>trabecular meshwork.10.4.4.1.3 Secondary OpenAngle GlaucomaParticulate or cellular elements present in <strong>the</strong> trabecularmeshwork can cause outflow obstruction. Examplesare iatrogenic glaucoma (caused by silicone oil, topicalsteroids or viscoelastic substances used to coat lens implants),haemolytic glaucoma, lens protein glaucoma,post-traumatic glaucoma <strong>and</strong> glaucoma in associationwith tumours (caused by necrotic cells <strong>of</strong> malignantmelanomas <strong>and</strong> retinoblastomas). The outflow systemcan also be blocked by melanin pigment granules releasedfrom iris stroma or pigment epi<strong>the</strong>lium when <strong>the</strong>iris is traumatised or becomes atrophic.10.4.4.1.4 Secondary AngleClosure GlaucomaIn secondary angle closure glaucoma, iridotrabecular oriridocorneal contact is present. This is most frequentlycaused by neovascular glaucoma, in which neovascularisationwith fibrosis <strong>of</strong> <strong>the</strong> iris occurs, for example inretinopathy <strong>of</strong> prematurity. O<strong>the</strong>r causes are end-stageinflammatory disease, retinal detachment, tumours ortrauma.10.4.4.2 CataractsProlonged exposure to ultraviolet light seems to be animportant cause <strong>of</strong> cataracts, a frequent disorder in elderlypatients. An extracted lens should be fixed, embeddedin paraffin wax <strong>and</strong> cut in two halves in <strong>the</strong> anteroposteriordirection. Slides can be made by sectioning <strong>the</strong>cut surface. Cutting <strong>the</strong> lens before processing can causeartefacts. In cataracts <strong>of</strong> <strong>the</strong> elderly degenerated lens fibresform discrete globules <strong>and</strong> <strong>the</strong> epi<strong>the</strong>lium covering<strong>the</strong> inner surface <strong>of</strong> <strong>the</strong> anterior lens capsule may extendto <strong>the</strong> posterior part <strong>of</strong> <strong>the</strong> lens. Similar observations aremade in cataracts in children [104].


298 M.R. Canninga-Van Dijk1010.4.4.3 Phtisis BulbiA long period <strong>of</strong> time after a trauma or an inflammatorydisease, <strong>the</strong> total eye becomes atrophic. As long as choroidal<strong>and</strong> retinal anatomy are preserved, this is calledatrophia bulbi. As soon as disorganisation <strong>of</strong> choroidea<strong>and</strong> retina occurs, it is called phtisis bulbi. A reactive cellproliferation dominates <strong>the</strong> histology. This proliferationcan be fibroblastic (trauma <strong>of</strong> cornea, sclera, choroideaor iris), or glial (retinal damage). Also, proliferation <strong>of</strong>retinal pigment epi<strong>the</strong>lium or ciliary body epi<strong>the</strong>liumcan be seen. The optic nerve is usually completely atrophic.10.4.4.4 Retinal Vascular DiseaseLoss <strong>of</strong> vision caused by ischaemic disease <strong>of</strong> <strong>the</strong> retinais common. It can be due to several different vasculardisorders. Most frequently it is caused by central retinalvein occlusion, diabetes, or occlusion <strong>of</strong> a branch vein.More rare are vasculitis, retinopathy <strong>of</strong> prematurity,radiation retinopathy, central retinal artery occlusion,hypertension <strong>and</strong> disseminated intravascular coagulopathy.Occlusion <strong>of</strong> an artery causes white infarction,while occlusion <strong>of</strong> veins leads to haemorrhagic infarction.The ischaemic area can vary between focal (occlusion<strong>of</strong> branch vessels), segmental, or total (occlusion <strong>of</strong><strong>the</strong> central retinal vein or artery). Ischaemia <strong>of</strong> <strong>the</strong> retinawith damage <strong>of</strong> retinal vasculature shows leakage <strong>of</strong>red cells, followed by neovascularisation <strong>and</strong> formation<strong>of</strong> microaneurysms. In <strong>the</strong> final stage, secondary angleclosure glaucoma can develop with corneal ulceration<strong>and</strong> cataract formation, resulting in a not only blind butalso painful eye. The globes are <strong>of</strong>ten enucleated to relievepain. At macroscopic examination an ectropion <strong>of</strong><strong>the</strong> iris pigment epi<strong>the</strong>lium, caused by <strong>the</strong> neovascularmembrane on <strong>the</strong> iris surface is visible (but only if <strong>the</strong>cornea is transparent). At microscopic examination, <strong>the</strong>proliferation <strong>of</strong> endo<strong>the</strong>lial cells in <strong>the</strong> retina is <strong>the</strong> moststriking finding. Sometimes CD31 <strong>and</strong> GFAP stainingare necessary to differentiate <strong>the</strong> vascular proliferationfrom reactive gliosis.10.4.4.5 Retinal DetachmentSeveral degenerative conditions predispose to retinal detachment.The separation between <strong>the</strong> neural retina <strong>and</strong><strong>the</strong> retinal pigment epi<strong>the</strong>lium can be caused by traction,exudate, or by so-called rhegmatogenous detachment.Traction detachment occurs when <strong>the</strong> vitreousshows fibrosis or gliosis, following trauma or by neovascularisation.Accumulation <strong>of</strong> fluid between <strong>the</strong> layers<strong>of</strong> <strong>the</strong> retina is called exudative detachment. It can becaused by processes with excessive permeability <strong>of</strong> retinalor choroidal vessels, like inflammatory or neoplasticdisorders. In rhegmatogenous detachment, passage <strong>of</strong>fluid from <strong>the</strong> vitreous cavity to <strong>the</strong> subretinal space ispresent. It occurs through a hole in <strong>the</strong> retina, caused bydegeneration or a minor trauma. Enucleated eyes withretinal detachment usually show many signs <strong>of</strong> previoussurgical intervention. The most important informationfor <strong>the</strong> surgeons is whe<strong>the</strong>r <strong>the</strong> retina survived <strong>the</strong> separation<strong>and</strong> reattachment or not <strong>and</strong> if a reason for surgicalfailure can be found.10.4.4.6 Retinitis PigmentosaRetinitis pigmentosa presents in early life with nightblindness <strong>and</strong> a progressive reduction in <strong>the</strong> visual field,starting at <strong>the</strong> periphery. Retinal architecture remainsbest preserved at <strong>the</strong> macula, so <strong>the</strong> patient ends up withtunnel vision. Different chromosomal abnormalities havebeen found in patients with retinitis pigmentosa. Microscopicexamination shows retinal atrophy with proliferation<strong>of</strong> Müller cells (retinal supporting cells at <strong>the</strong> outerside <strong>of</strong> <strong>the</strong> retina) replacing <strong>the</strong> outer nuclear layer. Theretinal pigment epi<strong>the</strong>lium proliferates <strong>and</strong> can surroundsmall hyalinised vessels in <strong>the</strong> retina. A marked variationin <strong>the</strong> extent <strong>of</strong> retinal degeneration can be seen in tworelatives with retinitis pigmentosa [117].10.4.5 Tumours<strong>and</strong> Tumour-Like Conditions10.4.5.1 MelanocyticMelanocytes in <strong>the</strong> uveal tract can give rise to both benign<strong>and</strong> malignant tumours. Racial differences mayreflect <strong>the</strong>mselves by variance in prominence <strong>and</strong> enlargement<strong>of</strong> melanocytes in <strong>the</strong> choroid, ciliary body<strong>and</strong> iris. It is very important for pathologists to be aware<strong>of</strong> <strong>the</strong>se differences.10.4.5.1.1 NaevusICD-O:8720/0Iris naevi present as pigmented macular lesions, arevery slowly progressive, <strong>and</strong> <strong>of</strong>ten completely static overyears. When <strong>the</strong> clinical presentation is not suspicious,in most cases <strong>the</strong> lesion will not be excised. For that reason,naevi <strong>of</strong> <strong>the</strong> uveal tract are most commonly incidentalfindings. Histology shows a symmetrical lesion,located in <strong>the</strong> anterior part <strong>of</strong> <strong>the</strong> iris stroma, <strong>and</strong> usuallycomposed <strong>of</strong> small spindle cells with small, uniformnuclei. Large nucleoli <strong>and</strong> especially mitotic figures indicatea suspected malignant melanoma.


Eye <strong>and</strong> Ocular Adnexa Chapter 10 299Fig. 10.19. Uveal tract melanoma arising in <strong>the</strong> choroidFig. 10.20. Uveal tract melanoma: detail <strong>of</strong> closely packed spindle-shapedcells; in this area almost no pigment is presentIn <strong>the</strong> ciliary body naevi are very rare; <strong>the</strong> histologyis comparable with iris naevi: spindle-shaped cellswithout atypia <strong>and</strong> without mitotic figures. In <strong>the</strong> choroid,naevus cells are spindle-shaped <strong>and</strong> <strong>of</strong>ten heavilypigmented, nuclei are uniform, <strong>and</strong> no mitotic figuresare seen. Depigmentation <strong>of</strong> <strong>the</strong> slides may be helpful inevaluating cytological details.10.4.5.1.2 Malignant MelanomaICD-O:8720/3The major part <strong>of</strong> uveal tract melanomas arise in <strong>the</strong>choroid (Fig. 10.19). Most frequently affected are whiteindividuals in <strong>the</strong> sixth <strong>and</strong> seventh decades <strong>of</strong> life.Most melanomas are single <strong>and</strong> confined to one eye.In most cases <strong>the</strong>y arise from pre-existing naevi in <strong>the</strong>choroid. Ciliary body <strong>and</strong> iris melanomas are rare.Malignant melanoma <strong>of</strong> <strong>the</strong> choroid presents with loss<strong>of</strong> vision in one eye, or with secondary closed angleglaucoma. The glaucoma is caused by detachment <strong>of</strong><strong>the</strong> retina with lens–pupil block. Spread <strong>of</strong> <strong>the</strong> tumourinto <strong>the</strong> orbita can cause proptosis. The primary clinicaldifferential diagnosis for melanoma is choroidalmetastasis. At macroscopic examination <strong>of</strong> an enucleatedeye with a choroidal melanoma, it is important tolocate <strong>the</strong> tumour before cutting <strong>the</strong> eye. The tumourcan be located by palpation <strong>and</strong> transillumination. Ifpossible, <strong>the</strong> main histological section should contain<strong>the</strong> centre <strong>of</strong> <strong>the</strong> pupil, <strong>the</strong> optic nerve <strong>and</strong> <strong>the</strong> centre<strong>of</strong> <strong>the</strong> tumour. Small pigmented nodules can be seenon <strong>the</strong> external surface <strong>of</strong> <strong>the</strong> sclera in case <strong>of</strong> transscleralextension <strong>of</strong> <strong>the</strong> tumour. The sample taken atthis point sometimes needs section at multiple levelsto demonstrate <strong>the</strong> tumour passing through <strong>the</strong> scleralcanals. Microscopically, <strong>the</strong> melanomas consist <strong>of</strong>Fig. 10.21. Uveal tract melanoma: a more pigmented areaspindle-shaped cells, epi<strong>the</strong>lioid cells, or a combination<strong>of</strong> both (mixed cell type). The spindle-shapedcells are closely packed elongated cells, <strong>of</strong>ten with pronouncednucleoli <strong>and</strong> a few mitotic figures. In epi<strong>the</strong>lioidlesions, <strong>the</strong> cytoplasm is more eosinophilic <strong>and</strong>mitotic figures are easily found. Melanin pigment isusually is present, but amelanotic lesions can be seen(Figs. 10.20–10.23). Positive immunohistochemistrywith S-100, melan A or HMB45 confirms <strong>the</strong> diagnosisin those cases. The final report should include <strong>the</strong>origin <strong>of</strong> <strong>the</strong> tumour (choroid, ciliary body, iris), <strong>the</strong>thickness <strong>of</strong> <strong>the</strong> tumour (in mm), <strong>the</strong> cell type (spindlecell type, epi<strong>the</strong>lioid cell type or mixed cell type) <strong>and</strong>extraocular growth. The 5-year survival <strong>of</strong> pure spindletumours is 80%, while pure epi<strong>the</strong>lioid tumourshave a 5-year survival <strong>of</strong> 35%. The 20-year survival <strong>of</strong>both groups is only 20%.


300 M.R. Canninga-Van DijkFig. 10.22. Uveal tract melanoma: detail <strong>of</strong> an epi<strong>the</strong>lioid lesionconsisting <strong>of</strong> cells with a large amount <strong>of</strong> eosinophilic cytoplasmFig. 10.24. Retinoblastoma: a detached retina with small blueround cell proliferation10Fig. 10.23. Uveal tract melanoma: trans-scleral extension <strong>of</strong> <strong>the</strong>tumour10.4.5.2 LymphoidIntraocular lymphomas are not different from those occuringelsewhere in <strong>the</strong> body. Therefore, discussion <strong>of</strong><strong>the</strong>ir features lies beyond <strong>the</strong> scope <strong>of</strong> this chapter.10.4.5.3 Retinoblastoma<strong>and</strong> PseudoretinoblastomaICD-O:9510/3Retinoblastoma is rare, but never<strong>the</strong>less <strong>the</strong> most commonintraocular malignant tumour <strong>of</strong> childhood, clinicallypresenting as a white mass behind <strong>the</strong> lens, resultingin <strong>the</strong> so-called cat’s eye reflex. Vision in <strong>the</strong> eye isimpaired, leading to strabismus. The tumour affectschildren younger than 5 years <strong>and</strong> can be unilateral orbilateral (30%). Retinoblastoma in adults is extremelyrare. Lesions with <strong>the</strong> typical clinical presentation butare <strong>of</strong> o<strong>the</strong>r kinds are called pseudoretinoblastoma. Theycan be o<strong>the</strong>r tumours (astrocytic hamartomas, haemangioblastomas),congenital malformations or inflammatoryconditions (especially solitary Toxocara granuloma).The accuracy <strong>of</strong> clinical diagnosis has been improvedby radiology, ultrasonography, CT scanning <strong>and</strong>nuclear magnetic resonance, <strong>and</strong> this has brought abouta considerable reduction in <strong>the</strong> number <strong>of</strong> such cases.The retinoblastoma gene, located on chromosome13q14 is a tumour-suppressor gene. Retinoblastoma canbe inherited (bilateral tumours in <strong>the</strong> first 2 years <strong>of</strong>life) or sporadic (unilateral tumours in children aged 2–5 years). One-third <strong>of</strong> <strong>the</strong> patients with a sporadic retinoblastomashow a germline mutation <strong>and</strong> can transmit<strong>the</strong> disease to <strong>the</strong>ir <strong>of</strong>fspring. Compared with <strong>the</strong> generalpopulation, carriers <strong>of</strong> germline mutations in <strong>the</strong> retinoblastomagene, who survive retinoblastoma are at increasedrisk <strong>of</strong> early-onset second cancers, particularlysarcomas <strong>and</strong> brain tumours. External beam radio<strong>the</strong>rapyhas been a st<strong>and</strong>ard treatment for medium <strong>and</strong>large, or vision-threatening, intraocular retinoblastoma,but it markedly increases <strong>the</strong> risk <strong>of</strong> cosmetic deformities<strong>and</strong> secondary cancer in children with germline mutations.For that reason primary systemic chemo<strong>the</strong>rapycalled “chemoreduction” has been employed to avoid radio<strong>the</strong>rapy<strong>and</strong> enucleation. The cure rate <strong>of</strong> retinoblastomais more than 90% in specialised centres.Retinoblastomas are tumours originating from pluripotentgerminal retinoblasts; <strong>the</strong> tumour can grow endophytically(growing into <strong>the</strong> vitreous), exophytically(growing into <strong>the</strong> subretinal space, leading to retinal detachment)or diffusely (a rare pattern with widespread


Eye <strong>and</strong> Ocular Adnexa Chapter 10 301From <strong>the</strong> glial tumours, only astrocytomas can be foundin <strong>the</strong> retina <strong>and</strong> optic nerve. Optic pathway gliomas arefrequently asymptomatic; sometimes <strong>the</strong>y demonstraterapid growth, causing considerable visual dysfunction,neurologic deficits, <strong>and</strong> endocrine disturbances. Mostoptic pathway gliomas are diagnosed in patients withneur<strong>of</strong>ibromatosis. Children with optic pathway gliomasassociated with neur<strong>of</strong>ibromatosis 1 predominantlyhave multifocal lesions.Benign astrocytic tumours <strong>of</strong> <strong>the</strong> retina ( astrocytichamartomas) most frequently occur in patients with tuberoussclerosis. Retinal astrocytic hamartoma <strong>and</strong> retinoblastomamay be very similar clinically <strong>and</strong> <strong>the</strong>ir differentiationin atypical cases can be difficult, even with<strong>the</strong> use <strong>of</strong> ultrasonography <strong>and</strong> computed tomography.Histologically, a well-circumscribed glial cell proliferation,sparing <strong>the</strong> outer layers <strong>of</strong> <strong>the</strong> retina will be visible.10.4.5.5 Vascular10.4.5.5.1 Angiomatosis RetinaeFig. 10.25. Retinoblastoma: detail <strong>of</strong> <strong>the</strong> small blue round cellswith high mitotic <strong>and</strong> apoptotic activitynodular thickening <strong>of</strong> <strong>the</strong> retina). The diffuse growthpattern has a bad prognosis. Trans-scleral spread <strong>of</strong> retinoblastomais uncommon. The tumour usually spreadsinto <strong>the</strong> meninges or parenchyma <strong>of</strong> <strong>the</strong> optic nerve. Forthis reason, it is important to take transverse blocks <strong>of</strong><strong>the</strong> cut surface <strong>of</strong> <strong>the</strong> optic nerve, before cutting <strong>the</strong> enucleatedeye. Microscopic examination will show a smallblue round cell tumour with a high mitotic rate. Thecells have ill-defined cytoplasm <strong>and</strong> inconspicuous nucleoli.Rosettes are frequently seen <strong>and</strong> apoptosis is common(Figs. 10.24, 10.25). Glial differentiation is rare. If<strong>the</strong> tumour was irradiated before enucleation, amorphouscalcified structures will be present. Immunohistochemistry<strong>of</strong> retinoblastomas will show positivity forS-100, GFAP <strong>and</strong> NSE. Use <strong>of</strong> <strong>the</strong>se markers can be helpfulnot only in identifying <strong>the</strong> tumour, but also in identifying<strong>the</strong> spread <strong>of</strong> <strong>the</strong> tumour. Choroidal invasion inparticular can be hard to recognise in an H&E staining.Axonal degeneration in <strong>the</strong> optic nerve, with reactiveproliferation <strong>of</strong> astrocytes has to be differentiated fromreal tumour spread.Von Hippel-Lindau disease is an autosomal dominantlyinherited multi-system disorder characterised by haemangioblasticlesions <strong>of</strong> <strong>the</strong> central nervous system <strong>and</strong>visceral organs [126]. Angiomatosis retinae (retinal haemangioblastoma)is <strong>of</strong>ten <strong>the</strong> first observable manifestation<strong>of</strong> von Hippel-Lindau disease. Histology showsa proliferation <strong>of</strong> capillary endo<strong>the</strong>lial cells <strong>and</strong> vacuolatedstromal cells.In some patients with von Hippel-Lindau disease orin <strong>the</strong> close relatives <strong>of</strong> such patients, unusual retinalvascular hamartomas o<strong>the</strong>r than retinal angiomas canbe detected.10.4.5.5.2 Cavernous<strong>and</strong> Capillary HaemangiomaICD-O:9120/0Haemangiomas <strong>of</strong> <strong>the</strong> choroid can be diagnosed clinicallyby fluorescein angiography. Diffuse haemangiomatosiswith facial skin involvement can be seen in <strong>the</strong>Sturge-Weber syndrome. The treatment <strong>of</strong> <strong>the</strong>se vascularlesions is radiation (external beam or radioactiveplaque); for this reason pathologists do not see <strong>the</strong>selesions very <strong>of</strong>ten. Only when <strong>the</strong> haemangiomas leadto retinal attachment <strong>and</strong> blindness will enucleation follow.The excised globe usually shows reactive changesdue to radio<strong>the</strong>rapy.10.4.5.4 Glial10.4.5.6 O<strong>the</strong>r Primary TumoursTumours o<strong>the</strong>r than melanocytic, lymphoid, retinoblastic<strong>and</strong> vascular can be found in <strong>the</strong> intraocular structures,but <strong>the</strong>y are extremely rare. In <strong>the</strong> iris leiomyoma,leiomyosarcoma, schwannoma, juvenile xanthogranuloma,rhabdomyosarcoma, inclusion cysts <strong>of</strong> <strong>the</strong> pigmentepi<strong>the</strong>lium, primary adenoma <strong>of</strong> <strong>the</strong> iris pigmentepi<strong>the</strong>lium <strong>and</strong> adenocarcinoma have been reported.In <strong>the</strong> ciliary body leiomyomas, leiomyosarcomas,schwannomas, adenomas <strong>and</strong> medulloepi<strong>the</strong>liomas can befound, <strong>and</strong> in <strong>the</strong> choroid osteomas, adenomas <strong>and</strong> adenocarcinomas<strong>of</strong> <strong>the</strong> retinal pigment epi<strong>the</strong>lium <strong>and</strong> hamar-


302 M.R. Canninga-Van Dijk10tomas have been described [63]. Choroidal osteomas c<strong>and</strong>evelop within a degenerated choroidal haemangioma oran inflammatory scar, but can also be idiopathic.10.4.5.7 Metastatic TumoursMetastatic tumours to <strong>the</strong> eye have a predilection for<strong>the</strong> highly vascular choroid. The metastases can be discoveredin a patient known to have a malignancy, but<strong>the</strong>y can also be <strong>the</strong> first presentation <strong>of</strong> <strong>the</strong> malignantdisease. Histology most <strong>of</strong>ten shows an adenocarcinoma<strong>and</strong> <strong>the</strong> primary tumour is found in <strong>the</strong> breast or lung[86]. More rare are metastases from thyroid carcinoma,carcinoid tumours, endometrial carcinoma, haemangiosarcoma<strong>and</strong> adenocarcinoma <strong>of</strong> <strong>the</strong> intestinal tract.The lesions usually show severe necrosis due to palliativeradio<strong>the</strong>rapy.10.5 Optic Nerve10.5.1 PapilloedemaAny condition in which <strong>the</strong> intracranial pressure israised can cause papilloedema. The prelaminar part <strong>of</strong><strong>the</strong> optic disc is swollen <strong>and</strong> <strong>the</strong> peripapillary photoreceptorsare placed laterally. If <strong>the</strong> reason for <strong>the</strong> papilloedemais not identified, <strong>the</strong> oedema can present as atumour. This so-called pseudotumour cerebri is treatedby optic nerve fenestration to relieve <strong>the</strong> pressure in <strong>the</strong>subarachnoid space. The pathologist will receive <strong>the</strong>meninges surrounding <strong>the</strong> optic nerve, which are histologicallycompletely normal.10.5.2 Optic NeuritisOptic neuritis presents as acute, unilateral <strong>and</strong> painfulvision loss. It can result from inflammatory disorders,can occur as isolated inflammation, or may be part <strong>of</strong><strong>the</strong> spectrum <strong>of</strong> multiple sclerosis. Magnetic resonanceimaging is indicated to rule out compressive optic neuropathy.Spontaneous outcome <strong>of</strong> optic neuropathiesis favourable. Secondary inflammatory optic neuritis(infection, vasculitis, sarcoidosis) is rare <strong>and</strong> usuallypresents with atypical evolution or o<strong>the</strong>r symptoms.Histologically, <strong>the</strong> optic nerve in multiple sclerosis willshow a perivascular lymphocytic infiltrate with focalareas <strong>of</strong> demyelination <strong>and</strong> axonal atrophy at <strong>the</strong> endstage.10.5.3 Optic AtrophyIn a normal optic disc, a large bulge <strong>of</strong> nerve fibre isformed. In enucleated glaucomatous eyes, <strong>the</strong> optic discis obviously cupped <strong>and</strong> shrinks down to <strong>the</strong> laminacribrosa, which becomes bowed posteriorly. Reactive fibrovasculartissue fills <strong>the</strong> cupped disc.10.5.4 Tumours10.5.4.1 GliomaICD-O:9380/3Most glial tumours <strong>of</strong> <strong>the</strong> optic nerve are <strong>of</strong> <strong>the</strong> juveniletype. They present with slowly progressive proptosis.The juvenile tumours have a good prognosis,unlike <strong>the</strong> more rare adult types, which are invariablylethal. Bilateral optic nerve gliomas can be found inpatients with neur<strong>of</strong>ibromatosis. Histologically, <strong>the</strong>juvenile tumours are <strong>of</strong> <strong>the</strong> pilocytic type, while <strong>the</strong>adult tumours resemble <strong>the</strong> high-grade glioblastomamultiforme.10.5.4.2 MeningiomaICD-O:9530/0Meningiomas surrounding <strong>the</strong> optic nerve can resultfrom a primary tumour arising in <strong>the</strong> meningeal sheet<strong>of</strong> <strong>the</strong> optic nerve or can result from an intracranialmeningioma spreading into <strong>the</strong> orbit. As in <strong>the</strong> intracranialcompartment, meningiomas <strong>of</strong> <strong>the</strong> orbit occurmost commonly in middle-aged females. The tumourspresent with proptosis. Optic nerve sheath meningiomasin <strong>the</strong> adult group grow slowly <strong>and</strong> have a goodprognosis. Meningiomas at o<strong>the</strong>r sites in <strong>the</strong> head <strong>and</strong>neck are mentioned in Chaps. 2, 6 <strong>and</strong> 8.10.6 Lacrimal Gl<strong>and</strong><strong>and</strong> Lacrimal Passages10.6.1 Inflammatory ProcessesEnlargement <strong>of</strong> <strong>the</strong> lacrimal gl<strong>and</strong> is <strong>of</strong>ten caused bychronic inflammatory processes that can have manydifferent causes. When chronic dacryoadenitis is associatedwith enlargement <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s, it iscalled Mikulicz syndrome. Mikulicz syndrome can becaused by many different diseases like sarcoidosis, tu-


Eye <strong>and</strong> Ocular Adnexa Chapter 10 303berculosis, mumps, malignant lymphoma <strong>and</strong> syphilis.The most frequent cause <strong>of</strong> Mikulicz syndromeis Mikulicz disease; <strong>the</strong> histology is similar to that <strong>of</strong>benign lymphoepi<strong>the</strong>lial lesions seen in <strong>the</strong> salivarygl<strong>and</strong>s. Acute <strong>and</strong> chronic dacryocystitis <strong>and</strong> canaliculitisare <strong>the</strong> result <strong>of</strong> inflammation, mostly nonspecific,<strong>of</strong> <strong>the</strong> lacrimal passages. It may lead to dacryolithiasis(stones in <strong>the</strong> lacrimal sac) <strong>and</strong> to lacrimalmucocele.10.6.2 Tumours<strong>and</strong> Tumour-Like ConditionsIf a mass is found in <strong>the</strong> superolateral quadrant <strong>of</strong> <strong>the</strong>orbit, dermoid cysts <strong>and</strong> lacrimal gl<strong>and</strong> masses shouldbe considered. Fifty percent <strong>of</strong> lacrimal gl<strong>and</strong> tumoursare pleomorphic adenomas <strong>and</strong> <strong>the</strong> o<strong>the</strong>r half are malignant[27, 98, 134]. The malignant category includes predominantlyadenoid cystic, carcinoma ex pleomorphicadenoma <strong>and</strong> mucoepidermoid carcinoma. The lacrimaltumours have few distinguishing imaging features,showing mostly a homogeneous character <strong>and</strong> moderatecontrast enhancement. Poorly defined margins withbone destruction suggest a malignancy, but even <strong>the</strong>malignant lesions can be relatively well defined. Thehistology <strong>of</strong> lacrimal gl<strong>and</strong> tumours is similar to that <strong>of</strong>salivary gl<strong>and</strong> tumours as discussed in Chap. 5. Papillomas<strong>and</strong> squamous cell carcinomas are <strong>the</strong> benign <strong>and</strong>malignant tumours most commonly seen in <strong>the</strong> lacrimalsac. Their microscopic features are similar to those arisingfrom <strong>the</strong> mucosa <strong>of</strong> <strong>the</strong> nose (see Chap. 2) or in <strong>the</strong>conjunctiva.10.7 Eyelids10.7.1 CystsBecause <strong>of</strong> <strong>the</strong> numerous adnexal gl<strong>and</strong>s present in <strong>the</strong>eyelids, cysts are very common at this localisation. Thecysts can be <strong>of</strong> developmental origin (dermoid cysts) orcan be caused by inclusion or retention.10.7.1.1 Dermoid CystThe most common type <strong>of</strong> a cyst <strong>of</strong> <strong>the</strong> eyelids in childrenis <strong>the</strong> dermoid cyst, a developmental cyst caused byinclusion <strong>of</strong> ectodermal rests within <strong>the</strong> lines <strong>of</strong> closure<strong>of</strong> <strong>the</strong> branchial arches. Dermoid cysts are lined withstratified squamous epi<strong>the</strong>lium with small pilosebaceousunits attached to <strong>the</strong> wall (Fig. 10.26). The lumenusually contains small hairs <strong>and</strong> keratin. The presenceFig. 10.26. Dermoid cyst: lined with squamous epi<strong>the</strong>lium withsmall pilosebaceous units in <strong>the</strong> wall<strong>of</strong> pilosebaceous units differentiates this cyst from epidermalcysts.10.7.1.2 Epidermal cystsEpidermal cysts (epidermoid cysts, keratinous cysts) arefirm, <strong>of</strong>ten yellow-brown masses, diagnosed clinicallyas “sebaceous cysts”. However, real sebaceous cysts (steatocystomas)are very rare <strong>and</strong> most cysts will histologicallyshow a lining with stratified squamous epi<strong>the</strong>liumwithout pilosebaceous gl<strong>and</strong>s. The cysts are filled withstr<strong>and</strong>s <strong>of</strong> keratin. Epidermal cysts can be caused bydermal inclusion <strong>of</strong> epi<strong>the</strong>lial cells after a microtrauma,but also by occlusion <strong>of</strong> a pilosebaceous unit. If an epidermalcyst ruptures, keratin will be released between<strong>the</strong> collagen bundles <strong>of</strong> <strong>the</strong> dermis, causing a granulomatousforeign body reaction.10.7.1.3 HidrocystomaCysts derived from <strong>the</strong> small sweat gl<strong>and</strong>s present in<strong>the</strong> eyelids present as bluish, round lesions <strong>and</strong> areclinically <strong>of</strong>ten misdiagnosed as haemangiomas. Theterm hidrocystoma is preferred, but many o<strong>the</strong>r names,like cysts <strong>of</strong> Moll’s gl<strong>and</strong>s <strong>and</strong> sudiferous cysts, can befound in <strong>the</strong> literature. The eccrine cysts are lined withcuboidal to flattened epi<strong>the</strong>lium with a myoepi<strong>the</strong>lialbase (Fig. 10.27). Sometimes only one epi<strong>the</strong>lial layeris visible, showing eosinophilic cytoplasm with snouts,characteristic <strong>of</strong> apocrine differentiation. It can be veryhard to differentiate between eccrine or apocrine origins<strong>and</strong> sometimes both components can be found in<strong>the</strong> cysts.


304 M.R. Canninga-Van DijkFig. 10.27. Hidrocystoma: cystic space lined with flattened epi<strong>the</strong>liumwith a myoepi<strong>the</strong>lial baseFig. 10.28. Chalazion: optical empty spaces surrounded by histiocytesforming granulomas. The infiltrate also contains lymphocytes<strong>and</strong> neutrophils1010.7.2 Inflammatory ProcessesA very common inflammatory condition <strong>of</strong> <strong>the</strong> eyelidsis a chalazion. Fur<strong>the</strong>rmore, many inflammatory skindiseases can involve <strong>the</strong> periorbital region. Periorbitaleczema may be an expression <strong>of</strong> a constitutional disease,an irritant or allergic dermatitis. O<strong>the</strong>r inflammatorydermatoses that can involve <strong>the</strong> eyelids are seborrheicdermatitis, psoriasis, rosacea <strong>and</strong> dermatomyositis.O<strong>the</strong>r causes <strong>of</strong> inflammation <strong>of</strong> <strong>the</strong> eyelids include bacterial,fungal <strong>and</strong> viral infections.10.7.2.1 Chalazion<strong>and</strong> O<strong>the</strong>r Ruptured CystsChalazia are very common. The clinical presentation isusually very typical, with an acute swelling in <strong>the</strong> tarsalconjunctiva. In a few days <strong>the</strong> swelling becomes afirm nodule. Excision or excochleation is <strong>the</strong> treatment<strong>of</strong> choice. Because <strong>of</strong> <strong>the</strong> typical clinical presentation,many ophthalmologists will not <strong>of</strong>fer <strong>the</strong> material forroutine histological examination. Chalazia are causedby <strong>the</strong> obstruction <strong>of</strong> <strong>the</strong> duct <strong>of</strong> a small (Zeis) or larger(Meibomian) sebaceous gl<strong>and</strong>. A small retention cyst isformed <strong>and</strong> rupture <strong>of</strong> this cyst causes <strong>the</strong> escape <strong>of</strong> fattyproducts into <strong>the</strong> surrounding tissues. The fatty materialtriggers an acute inflammatory reaction first, followedby a chronic granulomatous reaction (Fig. 10.28).In <strong>the</strong> very late stages <strong>of</strong> chalazia, fibrosis <strong>and</strong> scarringcan be seen. The presence <strong>of</strong> fatty cells or even largeroptical empty spaces within a granulomatous reaction ischaracteristic <strong>of</strong> a chalazion. The only o<strong>the</strong>r conditionswith similar lipogranulomatous reactions are leakage <strong>of</strong>implants (for example silicon) <strong>and</strong> dermatitis artefacta.If patients with dermatitis artefacta use oily fluids to inject,<strong>the</strong> histological picture is identical to that <strong>of</strong> a chalazion.All o<strong>the</strong>r cysts (dermoid, epidermal <strong>and</strong> hidrocystomas)can rupture. The wall <strong>of</strong> <strong>the</strong> cyst is sometimes nolonger identifiable. The presence <strong>of</strong> small pieces <strong>of</strong> <strong>the</strong>content <strong>of</strong> <strong>the</strong> former cyst (hair, keratin) in multinucleatedgiant cells proves <strong>the</strong> diagnosis <strong>of</strong> an inflamed <strong>and</strong>ruptured cyst.10.7.2.2 Deep Granuloma AnnulareGranuloma annulare usually occurs on <strong>the</strong> dorsum <strong>of</strong><strong>the</strong> h<strong>and</strong>s <strong>and</strong> <strong>the</strong> lower arms. It is considered to be acutaneous reaction pattern, most frequently associatedwith diabetes mellitus. However, in children <strong>the</strong> deepvariant <strong>of</strong> granuloma annulare is a benign, relativelycommon dermatosis, not related to systemic disease. Ingranuloma annulare <strong>of</strong> childhood lesions typically occuron <strong>the</strong> extremities <strong>and</strong> resolve spontaneously overa period <strong>of</strong> several months to years. Localised facial involvement,sometimes with involvement <strong>of</strong> <strong>the</strong> eyelids, israre. The clinical relevance is that granuloma annulare,presenting in <strong>the</strong> periocular region, may mimic o<strong>the</strong>rlesions. This diagnosis should be considered for any acquiredpapules <strong>of</strong> <strong>the</strong> periorbital area, especially if <strong>the</strong>reis a history <strong>of</strong> antecedent trauma. Unnecessary surgicalexcision can <strong>the</strong>n be avoided. Histology shows deep foci<strong>of</strong> degeneration <strong>of</strong> collagen, surrounded by histiocytes.Often <strong>the</strong>re is increased dermal mucin.


Eye <strong>and</strong> Ocular Adnexa Chapter 10 30510.7.2.3 Necrobiotic XanthogranulomaNecrobiotic xanthogranuloma is a rare chronic <strong>and</strong><strong>of</strong>ten progressive disorder with a predilection for <strong>the</strong>periorbital skin. O<strong>the</strong>r areas <strong>of</strong> <strong>the</strong> face, as well as <strong>the</strong>trunk <strong>and</strong> limbs, can also be involved. Lesions presentas sharply demarcated violaceus, partly xanthomatousnodules <strong>and</strong> plaques. Ulceration may develop. Almostall patients with necrobiotic xanthogranuloma are diagnosedwith a paraproteinaemia. O<strong>the</strong>r, more rare associationsare hyperlipidaemia <strong>and</strong> leukopenia. Scleritis,episcleritis <strong>and</strong> keratitis are common ophthalmiccomplications. The histological changes are present in<strong>the</strong> dermis <strong>and</strong> in <strong>the</strong> subcutis. Large zones <strong>of</strong> necrobioticcollagen with hyaline <strong>and</strong> sometimes mucinouschanges are present in <strong>the</strong> deep dermis. These areasare surrounded by histiocytes, partly with a foamycytoplasm. Sometimes <strong>the</strong> xanthomatous changesare only minor. Multinucleated giant cells are easilyfound; <strong>the</strong>y can be <strong>of</strong> <strong>the</strong> Touton type, but also <strong>of</strong> <strong>the</strong>foreign body type with bizarre nuclei. In ulceratinglesions, transepidermal elimination <strong>of</strong> debris can beseen.10.7.3 AmyloidosisSolitary or multiple nodules <strong>of</strong> amyloid may occur bothin <strong>the</strong> eyelids <strong>and</strong> in <strong>the</strong> conjunctiva. Most frequently itis a localised process, without signs <strong>of</strong> systemic amyloidosis.Histology shows amorphous, eosinophilic, Congored-positive masses in <strong>the</strong> stroma. The walls <strong>of</strong> bloodvessels <strong>of</strong>ten also contain amyloid.10.7.4 Tumours<strong>and</strong> Tumour-Like ConditionsTumours <strong>of</strong> <strong>the</strong> eyelids are very similar to tumours occurringin <strong>the</strong> conjunctiva <strong>and</strong> <strong>the</strong> skin. The most importanttumours <strong>of</strong> <strong>the</strong> eyelids are basal cell carcinomas,squamous cell carcinomas <strong>and</strong> sebaceous adenocarcinomas.These tumours are discussed in Sect. 10.2.6. Atumour-like condition with a predilection for <strong>the</strong> eyelids<strong>and</strong> <strong>the</strong> surrounding skin is xan<strong>the</strong>lasma.Fig. 10.29. Xan<strong>the</strong>lasmata: multiple foamy histiocytes are presentin <strong>the</strong> dermis10.8 Orbit10.8.1 Inflammatory ProcessesThe most common inflammatory diseases <strong>of</strong> <strong>the</strong> orbitinclude Graves’ disease (dysthyroid ophthalmopathy),orbital cellulitis <strong>and</strong> pseudotumours.10.8.1.1 Dysthyroid OphthalmopathyThe most common cause <strong>of</strong> bilateral proptosis is Graves’disease. Seventy percent <strong>of</strong> cases are bilateral <strong>and</strong> symmetrical.In cases <strong>of</strong> unilateral involvement, o<strong>the</strong>r diseasesmust be considered. Females are more frequentlyaffected than males. The disease is characterised by symmetricalswelling <strong>of</strong> <strong>the</strong> extraocular muscles. The inferior<strong>and</strong> medial rectus muscles are most <strong>of</strong>ten involved.The muscle enlargement characteristically involves <strong>the</strong>body <strong>of</strong> <strong>the</strong> muscle, sparing <strong>the</strong> tendinous attachment to<strong>the</strong> globe. Histologically, <strong>the</strong> fibrous tissue <strong>of</strong> <strong>the</strong> orbit<strong>and</strong> <strong>the</strong> swollen muscles show oedema <strong>and</strong> chronic inflammationin early stages <strong>and</strong> fibrosis in end-stage disease.The degenerated muscle fibres become hyalinised.10.7.4.1 Xan<strong>the</strong>lasmataXan<strong>the</strong>lasmata are yellow papules <strong>and</strong> plaques, most frequentlyoccurring on <strong>the</strong> eyelids <strong>and</strong> <strong>the</strong> skin surrounding<strong>the</strong> eyes. They are relatively frequent. Xan<strong>the</strong>lasmatacan be a manifestation <strong>of</strong> hypercholesterolaemia,but most cases are idiopathic. Histology shows multiplefoamy histiocytes in <strong>the</strong> superficial dermis (Fig. 10.29).10.8.1.2 CellulitisAcute bacterial infection (cellulitis) <strong>of</strong> <strong>the</strong> orbit is uncommon.It is most frequently caused by direct spread<strong>of</strong> an infection from <strong>the</strong> paranasal sinuses or eyelids. Itmay also be <strong>of</strong> odontogenic origin <strong>and</strong> can be one <strong>of</strong> <strong>the</strong>presenting features <strong>of</strong> retinoblastoma or o<strong>the</strong>r tumours.Chronic intranasal cocaine abuse can result in exten-


306 M.R. Canninga-Van Dijk<strong>and</strong> fat in <strong>the</strong> earliest stages. This will be followed bylymphocytic infiltration <strong>and</strong> end with fibrous changes.In cases <strong>of</strong> massive infiltration by lymphocytes, immunohistochemistryis necessary to rule out malignantlymphoma (Fig. 10.30). Ano<strong>the</strong>r disease that canmimic pseudotumours is sarcoidosis. Sarcoidosis ishistologically characterised by typical granulomas, notsurrounded by lymphocytes.10Fig. 10.30. Pseudotumour <strong>of</strong> <strong>the</strong> orbit: fibrous tissue with necroticfat cells is infiltrated by large groups <strong>of</strong> lymphocytes. Immunohistochemistryis necessary to rule out a malignant lymphomasive bony destruction <strong>of</strong> <strong>the</strong> orbital walls with associatedorbital cellulitis. In patients with poorly controlleddiabetes, but also in immunocompromised patients,orbital cellulitis can also be caused by fungal agents,for example mucormycosis. Presenting symptoms mostfrequently include oedema <strong>of</strong> <strong>the</strong> upper eyelid, headache<strong>and</strong> facial pain. Sometimes it can be asymptomatic.Clinically, orbital cellulitis is <strong>of</strong> great importance,as it is a severe disease with potentially disastrous consequences.Despite antifungal or antibacterial <strong>the</strong>rapy,disease can progress. It may lead to optic neuritis, opticatrophy, blindness, cavernous sinus thrombosis, intracranialabscess formation, meningitis, subdural empyema,<strong>and</strong> even death. An incision biopsy <strong>of</strong> <strong>the</strong> processcan be helpful in <strong>the</strong> diagnostic work-up. Histology willshow an extensive neutrophilic infiltration <strong>of</strong> <strong>the</strong> orbitalfibrous tissue <strong>and</strong> fat. The causative microorganismscan <strong>of</strong>ten be found with PAS, Gram <strong>and</strong> silver stainings.It is important for <strong>the</strong> pathologist to look for underlyingcauses, like tumours.10.8.1.3 PseudotumourNon-specific inflammation <strong>of</strong> orbital tissues is knownas orbital pseudotumour. It tends to be unilateral <strong>and</strong>accounts for 25% <strong>of</strong> all cases <strong>of</strong> unilateral exophthalmos.Spontaneous regression can occur <strong>and</strong> a responseto steroids is <strong>of</strong>ten seen. However, orbital pseudotumourscan also be chronic <strong>and</strong> progressive. The diagnosishas to be confirmed by an incision biopsy, especiallyin cases in which <strong>the</strong> pseudotumour appearsas a discrete mass <strong>and</strong> simulates a neoplastic lesion.Histology shows oedema <strong>of</strong> <strong>the</strong> orbital fibrous tissue10.8.2 Tumours<strong>and</strong> Tumour-Like ConditionsA variety <strong>of</strong> tumours <strong>and</strong> pseudotumours can involve<strong>the</strong> orbit. Most orbital lesions are benign (65%). The percentage<strong>of</strong> malignant tumours increases with age, with60% <strong>of</strong> malignancies in patients over 60 years <strong>of</strong> age, because<strong>of</strong> <strong>the</strong> higher incidence <strong>of</strong> lymphoma <strong>and</strong> metastatictumours in <strong>the</strong> elderly. Orbital tumours <strong>of</strong> childhoodare distinct from tumours that occur in adults. Manyare congenital with early presentations. Most paediatricorbital tumours are benign (80%); developmental cystscomprise half <strong>of</strong> orbital cases, with capillary haemangiomabeing <strong>the</strong> second most common orbital tumourin children. The most common orbital malignancy inchildren is rhabdomyosarcoma. Whereas <strong>the</strong> malignanttumours may be life-threatening, both malignant <strong>and</strong>benign tumours may be vision-threatening. Almost alllymphomas, s<strong>of</strong>t tissue <strong>and</strong> bone tumours may involve<strong>the</strong> orbit.10.8.2.1 Developmental CystsEpi<strong>the</strong>lial rests found at sutural sites within <strong>the</strong> orbitcan give rise to epi<strong>the</strong>lial cysts. Cysts <strong>of</strong> <strong>the</strong> surfaceepi<strong>the</strong>lium are fur<strong>the</strong>r divided into simple epi<strong>the</strong>lialcysts (epidermal, conjunctival, respiratory <strong>and</strong> apocrinegl<strong>and</strong>), <strong>and</strong> dermoid cysts (epidermal <strong>and</strong> conjunctival).Epidermal dermoid cyst (dermoid) is by far <strong>the</strong> mostcommon orbital cystic lesion in children, accountingfor over 40% <strong>of</strong> all orbital lesions <strong>of</strong> childhood. O<strong>the</strong>rdevelopmental cysts are teratomatous cysts, neural cysts(congenital cystic eye <strong>and</strong> colobomatous cyst) <strong>and</strong> thoseassociated with brain <strong>and</strong> meningeal tissue (encephalocele<strong>and</strong> optic nerve meningocele) [103]. Developmentalcysts have to be differentiated from secondary cysts, likemucocele <strong>and</strong> inflammatory cysts. Mucocele can occurin children with cystic fibrosis. Inflammatory cystsare generally due to parasitic infestations <strong>and</strong> are morecommon in tropical areas <strong>of</strong> <strong>the</strong> world. Fur<strong>the</strong>rmore,non-cystic tumourous lesions with a cystic component(like rhabdomyosarcoma <strong>and</strong> lymphangioma) can presentas a cyst.


Eye <strong>and</strong> Ocular Adnexa Chapter 10 307The orbit is <strong>the</strong> most common location for metastasesto <strong>the</strong> eye <strong>and</strong> adjacent structures in children (neuroblastoma),whereas <strong>the</strong> choroid is <strong>the</strong> predominant sitein adults. Approximately 5% <strong>of</strong> all orbital tumour-likelesions are metastatic lesions. However, because malignanciesare far more frequent in adults, most orbitalmetastatic lesions are found in elderly patients. Themean period <strong>of</strong> time between <strong>the</strong> onset <strong>of</strong> <strong>the</strong> primarydisease <strong>and</strong> orbital manifestation is 5 years. The mainprimary symptoms are lid swelling, red eye, diplopia <strong>and</strong>proptosis. The most frequent primary tumour is a breastcarcinoma, but many o<strong>the</strong>r carcinomas can metastasiseto <strong>the</strong> orbit (Figs. 10.31, 10.32). Metastatic melanomas to<strong>the</strong> eye <strong>and</strong> orbit are rare <strong>and</strong> generally occur in patientswith disseminated metastases during <strong>the</strong> terminal stages<strong>of</strong> <strong>the</strong> disease, with a short life expectancy.Fig. 10.31. Carcinoid metastatic to <strong>the</strong> orbit: this haemorrhagicorbital mass contained small nests <strong>of</strong> monomorphous epi<strong>the</strong>lialcells. Immunohistochemistry was consistent with a carcinoidFig. 10.32. Renal cell adenocarcinoma metastatic to <strong>the</strong> orbit:nests <strong>of</strong> clear epi<strong>the</strong>lioid cells were found; immunohistochemistryshowed positivity for both vimentin <strong>and</strong> cytokeratin10.8.2.2 Optic Nerve<strong>and</strong> Meningeal TumoursOptic nerve <strong>and</strong> meningeal tumours can spread into<strong>the</strong> orbit. Toge<strong>the</strong>r <strong>the</strong>y represent 8% <strong>of</strong> all orbital tumours.10.8.2.3 Metastatic TumoursReferences1. Akpek EK, Polcharoen W, Chan R, Foster CS (1999) Ocularsurface neoplasia masquerading as chronic blepharoconjunctivitis.Cornea 18:282–2882. Akpek EK, Polcharoen W, Ferry JA, Foster CS (1999) Conjunctivallymphoma masquerading as chronic conjunctivitis.Ophthalmology 106:757–7603. Armstrong NT (1992) The ocular manifestations <strong>of</strong> congenitalrubella syndrome. Insight 17:14–164. Aultbrinker EA, Starr MB, Donnenfeld ED (1988) Linear IgAdisease. The ocular manifestations. Ophthalmology 95:340–3435. Balayre S, Gomez K, Tribut A, Dore P, Gobert F (2003) Toxoplasmagondii <strong>and</strong> necrotizing retinitis: a case report. J FrOphtalmol 26:837–8416. Bastiaensen LA, Verpalen MC, Pijpers PM, Sprong AC (1985)Conjunctival sarcoidosis. Doc Ophthalmol 59:5–97. Biggs SL, Font RL (1977) Oncocytic lesions <strong>of</strong> <strong>the</strong> caruncle<strong>and</strong> o<strong>the</strong>r ocular adnexa. Arch Ophthalmol 95:474–4788. Bost M, Mouillon M, Romanet JP, Deiber M, Navoni F (1985)Congenital glaucoma <strong>and</strong> Von Recklinghausen’s disease. Pediatrie40:207–2129. Bourcier T, Monin C, Baudrimont M, Larricart P, Borderie V,Laroche L (2003) Conjunctival inclusion cyst following parsplana vitrectomy. Arch Ophthalmol 121:106710. Breslow NE, Norris R, Norkool PA, Kang T, Beckwith JB,Perlman EJ, Ritchey ML, Green DM, Nichols KE (2003)Characteristics <strong>and</strong> outcomes <strong>of</strong> children with <strong>the</strong> Wilms tumor-Aniridiasyndrome: a report from <strong>the</strong> National WilmsTumor Study Group. J Clin Oncol 21:4579–458511. Bruna F (1954) Association <strong>of</strong> keratoconus with retinitis pigmentosa.Boll Ocul 33:145–15712. Busbee BG (2004) Advances in knowledge <strong>and</strong> treatment: anupdate on endophthalmitis. Curr Opin Ophthalmol 15:232–23713. Cervantes G, Rodriguez AA Jr, Leal AG (2002) Squamouscell carcinoma <strong>of</strong> <strong>the</strong> conjunctiva: clinicopathological featuresin 287 cases. Can J Ophthalmol 37:14–19; discussion19–2014. Ceuterick C, Martin JJ (1984) Diagnostic role <strong>of</strong> skin or conjunctivalbiopsies in neurological disorders. An update. JNeurol Sci 65:179–19115. Chai F, Coates H (2003) Otolaryngological manifestations<strong>of</strong> ligneous conjunctivitis. Int J Pediatr Otorhinolaryngol67:189–19416. Chan CC, BenEzra D, Hsu SM, Palestine AG, NussenblattRB (1985) Granulomas in sympa<strong>the</strong>tic ophthalmia <strong>and</strong> sarcoidosis.Immunohistochemical study. Arch Ophthalmol103:198–202


308 M.R. Canninga-Van Dijk1017. Chen S, Wishart M, Hiscott P (2000) Ligneous conjunctivitis:a local manifestation <strong>of</strong> a systemic disorder? J AAPOS4:313–31518. Cibis GW (1987) Congenital glaucoma. J Am Optom Assoc58:728–73319. Cibis GW, Tripathi RC, Tripathi BJ (1984) Glaucoma inSturge-Weber syndrome. Ophthalmology 91:1061–107120. Claes K, Kestelyn P (2000) Ocular manifestations <strong>of</strong> graftversus host disease following bone marrow transplantation.Bull Soc Belge Ophtalmol 21–2621. Cogan DG (1971) Congenital anomalies <strong>of</strong> <strong>the</strong> retina. BirthDefects Orig Artic Ser 7:41–5122. Craitoiu S (1992) Goldenhar’s oculoauricular dysplasia, limbicdermoid <strong>and</strong> conjunctival dermolipoma. Oftalmologia36:357–36123. Crolla JA, van Heyningen V (2002) Frequent chromosomeaberrations revealed by molecular cytogenetic studies in patientswith aniridia. Am J Hum Genet 71:1138–114924. Cursiefen C, H<strong>of</strong>mann-Rummelt C, Schlotzer-Schrehardt U,Fischer DC, Kuchle M (2000) Immunophenotype classification<strong>of</strong> macular corneal dystrophy: first case report <strong>of</strong> immunophenotypeI A outside <strong>of</strong> Saudi Arabia. A clinical histopathologicalcorrelation with immunohistochemistry <strong>and</strong>electron microscopy. Klin Monatsbl Augenheilkd 217:118–12625. Darougar S, Pearce R, Gibson JA, McSwiggan DA (1978)Adenovirus type 21 keratoconjunctivitis. Br J Ophthalmol62:836–83726. Dart J (2003) Corneal toxicity: <strong>the</strong> epi<strong>the</strong>lium <strong>and</strong> stroma iniatrogenic <strong>and</strong> factitious disease. Eye 17:886–89227. De Rosa G, Zeppa P, Tranfa F, Bonavolonta G (1986) Aciniccell carcinoma arising in a lacrimal gl<strong>and</strong>. First case report.Cancer 57:1988–199128. Dokonalova E, Vymazalova Z, Vymazal M, Riegrova D(1990) A complex choristoma <strong>of</strong> <strong>the</strong> conjunctiva. Acta UnivPalacki Olomuc Fac Med 126:227–23129. El-Ashry MF, El-Aziz MM, Larkin DF, Clarke B, Cree IA,Hardcastle AJ, Bhattacharya SS, Ebenezer ND (2003) A clinical,histopathological, <strong>and</strong> genetic study <strong>of</strong> Avellino cornealdystrophy in British families. Br J Ophthalmol 87:839–84230. Elsas FJ, Green WR (1975) Epibulbar tumors in childhood.Am J Ophthalmol 79:1001–100731. Faraj HG, Hoang-Xuan T (2001) Chronic cicatrizing conjunctivitis.Curr Opin Ophthalmol 12:250–25732. Fern<strong>and</strong>es M, Vemuganti GK, Pasricha G, Bansal AK, SangwanVS (2003) Unilateral tuberculous conjunctivitis withtarsal necrosis. Arch Ophthalmol 121:1475–147833. Freedman J, Gombos GM (1971) Bilateral macular coloboma,keratoconus, <strong>and</strong> retinitis pigmentosa. Ann Ophthalmol3:664–66534. Gamel JW, Eiferman RA, Guibor P (1984) Mucoepidermoidcarcinoma <strong>of</strong> <strong>the</strong> conjunctiva. Arch Ophthalmol 102:730–73135. Gayre GS, Proia AD, Dutton JJ (2002) Epibulbar osseouschoristoma: case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. OphthalmicSurg Lasers 33:410–41536. Gloor P, Ansari I, Sinard J (1999) Sebaceous carcinoma presentingas a unilateral papillary conjunctivitis. Am J Ophthalmol127:458–45937. Haller EM, Auer-Grumbach P, Stuenzner D, Kessler HH,Pierer K, Zenz H, Muellner K (1996) Evaluation <strong>of</strong> two noncultureantigen tests <strong>and</strong> three serotests for detection <strong>of</strong> antichlamydialantibodies in <strong>the</strong> diagnosis <strong>of</strong> ocular chlamydialinfections. Graefes Arch Clin Exp Ophthalmol 234:510–51438. Hanna C (1976) Embryopathic time-table <strong>of</strong> cataract formationin congenital rubella. J Pediatr Ophthalmol 13:266–27339. Hegab SM, al-Mutawa SA, Sheriff SM (1998) Sarcoidosis presentingas multilobular limbal corneal nodules. J PediatrOphthalmol Strabismus 35:323–32640. Heiligenhaus A, Dutt JE, Foster CS (1996) Histology <strong>and</strong> immunopathology<strong>of</strong> systemic lupus ery<strong>the</strong>matosus affecting<strong>the</strong> conjunctiva. Eye 10:425–43241. Ho MW, Crean SJ, Sharma A, Sukhija J, Das A, Saroha V,Sukhi S, Mohan K (2003) Simultaneous occurrence <strong>of</strong> sublingualdermoid cyst <strong>and</strong> oral alimentary tract cyst in an infant:a case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. Int J PaediatrDent 13:441–446.42. Ho VT, Rao VM, Fl<strong>and</strong>ers AE (1994) Postsurgical conjunctivalepi<strong>the</strong>lial cysts. AJNR Am J Neuroradiol 15:1181–118343. Hochman MA, Mayers M (1997) Stevens-Johnson syndrome,epidermolysis bullosa, staphylococcal scalded skin syndrome,<strong>and</strong> dermatitis herpetiformis. Int Ophthalmol Clin37:77–9244. Jakobiec FA, Folberg R, Iwamoto T (1989) Clinicopathologiccharacteristics <strong>of</strong> premalignant <strong>and</strong> malignant melanocyticlesions <strong>of</strong> <strong>the</strong> conjunctiva. Ophthalmology 96:147–16645. Jordaan HF, Schneider JW, Schaaf HS, Victor TS, Geiger DH,Van Helden PD, Rossouw DJ (1996) Papulonecrotic tuberculidin children. A report <strong>of</strong> eight patients. Am J Dermatopathol18:172–18546. Kallor AD, Hendricks WM, Salvitti C, Azulay RD, HeringerML, Almeida de Faria IA (1979) Oculoauriculovertebral dysplasia(Goldenhar’s syndrome). Cutis 24:41-4247. Karcioglu ZA, Issa TM (1997) Human papilloma virus inneoplastic <strong>and</strong> non-neoplastic conditions <strong>of</strong> <strong>the</strong> external eye.Br J Ophthalmol 81:595–59848. Kawamoto K, Miyanaga Y, Toriyama S (1996) A case <strong>of</strong> bilateralMALT (mucosa-associated lymphoid tissue) typeconjunctival malignant lymphoma. Nippon Ganka GakkaiZasshi 100:246–25249. Kaye SB, Baker K, Bonshek R, Maseruka H, Grinfeld E, TulloA, Easty DL, Hart CA (2000) Human herpesviruses in <strong>the</strong>cornea. Br J Ophthalmol 84:563–57150. Kerty E, Vig<strong>and</strong>er K, Flage T, Brinch L (1999) Ocular findingsin allogeneic stem cell transplantation without totalbody irradiation. Ophthalmology 106:1334–133851. Kh<strong>and</strong>ekar R, Al Awaidy S, Ganesh A, Bawikar S (2004) Anepidemiological <strong>and</strong> clinical study <strong>of</strong> ocular manifestations<strong>of</strong> congenital rubella syndrome in Omani children. ArchOphthalmol 122:541–54552. Kim JH, Hwang JM, Kim HJ, Yu YS (2002) Characteristic ocularfindings in Asian children with Down syndrome. Eye16:710–71453. Knoll A (1955) Simultaneous occurrence <strong>of</strong> degenerative retinitispigmentosa <strong>and</strong> keratoconus. Szemeszet 92:165–16854. Korvatska E, Munier FL, Djemai A, Wang MX, Frueh B,Chiou AG, Uffer S, Ballestrazzi E, Braunstein RE, ForsterRK, Culbertson WW, Boman H, Zografos L, Schorderet DF(1998) Mutation hot spots in 5q31-linked corneal dystrophies.Am J Hum Genet 62:320–32455. Kruk J, Kallor AD, Hendricks WM, Salvitti C, Azulay RD,Heringer ML, Almeida de Faria IA (1979) Goldenhar’s syndrome:aetiopathogenesis, morphology <strong>and</strong> treatment. KlinOczna 81:485–48656. Kuming BS, J<strong>of</strong>fe L (1977) Ehlers-Danlos syndrome associatedwith keratoconus. A case report. S Afr Med J 52:403–40557. Kunimoto DY, Tasman W, Rapuano C, Recchia F, Busbee B,Pearlman R, Belmont J, Cohen E, V<strong>and</strong>er J, Laibson P, RaberI (2004) Endophthalmitis after penetrating keratoplasty: microbiologicspectrum <strong>and</strong> susceptibility <strong>of</strong> isolates. Am JOphthalmol 137:343–34558. Lam S, Stone MS, Goeken JA, Massicotte SJ, Smith AC, FolbergR, Krachmer JH (1992) Paraneoplastic pemphigus, cicatricialconjunctivitis, <strong>and</strong> acanthosis nigricans with pachydermatoglyphyin a patient with bronchogenic squamous cellcarcinoma. Ophthalmology 99:108–11359. Langlois M, Evain B, Fournier E, Potier F (1986) Choroidalpseudotumoral granuloma in sarcoidosis (apropos <strong>of</strong> a case).Bull Soc Ophtalmol Fr 86:223–22460. Lesser RL (1995) Ocular manifestations <strong>of</strong> Lyme disease. AmJ Med 98:60S–62S61. Liesegang TJ (1994) Pigmented conjunctival <strong>and</strong> scleral lesions.Mayo Clin Proc 69:151–161


Eye <strong>and</strong> Ocular Adnexa Chapter 10 30962. Lin J, Li Y, Zhang J, Feng G, Zhang P, Zheng H, Zheng J(1999) Rapid diagnosis <strong>of</strong> chlamydial conjunctivitis in laboratory.Yan Ke Xue Bao 15:191–19463. Liu CC, Kou HK, Tsai MH, Su CC, Kao ML, Chen YJ, TsaiSH (2001) Choroidal masses: a fourteen-year analysis. ChangGung Med J 24:502–51164. Livesey SJ, Holmes JA, Whittaker JA (1989) Ocular complications<strong>of</strong> bone marrow transplantation. Eye 3:271–27665. Lloyd IC, Colley A, Tullo AB, Bonshek R (1993) Dominantlyinherited unilateral retinal dysplasia. Br J Ophthalmol77:378–38066. Longanesi L, Cavallini GM, Iammarino A, Vaccina F, GuerraR, De Pol A (1998) Ultrastructural localization <strong>of</strong> gelsolin inlattice corneal dystrophy type I. Ophthalmologica 212:415–42167. Luo Q, Wang L, Feng X, Cai R, Shen G, Deng Y, Xia R (1999)Observation <strong>of</strong> special stain <strong>and</strong> ultrastructure <strong>of</strong> lattice,granular <strong>and</strong> macular corneal dystrophy. Zhonghua Yan KeZa Zhi 35:268–27068. MacCallan A (1931) The epidemiology <strong>of</strong> trachoma. Br JOphthalmol 15:369–41169. Martin DF, Chan CC, de Smet MD, Palestine AG, Davis JL,Whitcup SM, Burnier MN Jr, Nussenblatt RB (1993) The role<strong>of</strong> chorioretinal biopsy in <strong>the</strong> management <strong>of</strong> posterior uveitis.Ophthalmology 100:705–71470. Mauriello JA Jr, Abdelsalam A, McLean IW (1997) Adenoidsquamous carcinoma <strong>of</strong> <strong>the</strong> conjunctiva – a clinicopathologicalstudy <strong>of</strong> 14 cases. Br J Ophthalmol 81:1001–100571. McDonnell JM, McDonnell PJ, Sun YY (1992) Human papillomavirusDNA in tissues <strong>and</strong> ocular surface swabs <strong>of</strong> patientswith conjunctival epi<strong>the</strong>lial neoplasia. Invest OphthalmolVis Sci 33:184–18972. Merchant S, Weinstein M (2003) Pemphigus vulgaris: <strong>the</strong>eyes have it. Pediatrics 112:183–18573. Messmer EM, Foster CS (1999) Vasculitic peripheral ulcerativekeratitis. Surv Ophthalmol 43:379–39674. Meyers SJ, Varley GA, Meisler DM, Camisa C, W<strong>and</strong>er AH(1992) Conjunctival involvement in paraneoplastic pemphigus.Am J Ophthalmol 114:621–62475. Mohile M, Deorari AK, Satpathy G, Sharma A, Singh M(2002) Microbiological study <strong>of</strong> neonatal conjunctivitis withspecial reference to Chlamydia trachomatis. Indian J Ophthalmol50:295–29976. Mubarik M, Nabi B, Ladakhi GM, Sethi AS (2000) Childhoodtuberculosis. I. Epidemiology, pathogenesis, clinicalpr<strong>of</strong>ile. JK Pract 7:12–1577. Munier FL, Korvatska E, Djemai A, Le Paslier D, Zografos L,Pescia G, Schorderet DF (1997) Kerato-epi<strong>the</strong>lin mutationsin four 5q31-linked corneal dystrophies. Nat Genet 15:247–25178. Neumann R, Dutt CJ, Foster CS (1993) Immunohistopathologicfeatures <strong>and</strong> <strong>the</strong>rapy <strong>of</strong> conjunctival lichen planus. AmJ Ophthalmol 115:494–50079. Oehme A, Musholt PB, Dreesbach K (1991) Chlamydiae aspathogens – an overview <strong>of</strong> diagnostic techniques, clinicalfeatures, <strong>and</strong> <strong>the</strong>rapy <strong>of</strong> human infections. Klin Wochenschr69:463–47380. Oguz O, Gokler G, Ocakoglu O, Oguz V, Demirkesen C,Aydemir EH (1997) Conjunctival involvement in familialchronic benign pemphigus (Hailey-Hailey disease). Int JDermatol 36:282–28581. O’Neal ML, Brunson A, Spadafora J (2001) Ocular sebaceouscarcinoma: case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. ComprTher 27:144–14782. Palazzi MA, Erwenne CM, Villa LL (2000) Detection <strong>of</strong> humanpapillomavirus in epi<strong>the</strong>lial lesions <strong>of</strong> <strong>the</strong> conjunctiva.Sao Paulo Med J 118:125–13083. P<strong>and</strong>a A, Arora R, Mohan M (1987) Cystic lesions <strong>of</strong> conjunctiva.Ann Ophthalmol 19:60–6284. Payne MS, Nadell JM, Lacassie Y, Tilton AH (2003) Congenitalglaucoma <strong>and</strong> neur<strong>of</strong>ibromatosis in a monozygotictwin: case report <strong>and</strong> review <strong>of</strong> <strong>the</strong> literature. J Child Neurol18:504–50885. Pleyer U, Bergmann L, Krause A, Hartmann C (1996) Autoimmunediseases <strong>of</strong> <strong>the</strong> peripheral cornea. Immunopathology,clinical aspects <strong>and</strong> <strong>the</strong>rapy. Klin Monatsbl Augenheilkd208:73–8186. Potter De P (1998) Ocular manifestations <strong>of</strong> cancer. CurrOpin Ophthalmol 9:100–10487. Pouliquen Y, Giraud JP, Savoldelli M (1978) Reis-Buckler’sdystrophy. Albrecht Von Graefes Arch Klin Exp Ophthalmol208:25–3188. Rabinowitz YS (1998) Keratoconus. Surv Ophthalmol42:297–31989. Ravault MP, Trepsat C, Chatenoud F (1983) C<strong>and</strong>ida albicansendophthalmitis in drug addicts: apropos <strong>of</strong> 2 cases. Bull SocOphtalmol Fr 83:1307–130890. Reccia R, Del Prete A, Scala A, Masciello M, Di Giovanni A,Sebastiani A (1994) Direct immun<strong>of</strong>luorescence <strong>and</strong> scrapingconjunctival cytology in <strong>the</strong> study <strong>of</strong> 912 patients affectedby micr<strong>of</strong>ollicular conjunctivitis. Ophthalmologica 208:295–29791. Reedy EA (2004) The discovery <strong>of</strong> retrolental fibroplasia <strong>and</strong><strong>the</strong> role <strong>of</strong> oxygen: a historical review, 1942–1956. NeonatalNetw 23:31–3892. Riley GP, Harrall RL, Watson PG, Cawston TE, HazlemanBL (1995) Collagenase (MMP-1) <strong>and</strong> TIMP-1 in destructivecorneal disease associated with rheumatoid arthritis. Eye9:703–71893. Ring CC (1958) Atopic dermatitis with cataracts; a report <strong>of</strong>three cases, one with keratoconus. Trans Ophthalmol Soc NZ 10:41–4794. Robertson I (1975) Keratoconus <strong>and</strong> <strong>the</strong> Ehlers-Danlos syndrome:a new aspect <strong>of</strong> keratoconus. Med J Aust 1:571–57395. Rosenberg ME, Tervo TM, Gallar J, Acosta MC, Muller LJ,Moilanen JA, Tarkkanen AH, Vesaluoma MH (2001) Cornealmorphology <strong>and</strong> sensitivity in lattice dystrophy type II(familial amyloidosis, Finnish type). Invest Ophthalmol VisSci 42:634–64196. Ryckewaert M, Zanlonghi X, Castier P, Francois P (1988)Choroidal granuloma in sarcoidosis. Discussion apropos <strong>of</strong>2 cases. J Fr Ophtalmol 11:773–77897. Sanchez-Tocino H, Saornil MA, Herreras JM, Blanco G,Calonge M, Rodriguez De La Rua E (2001) Usefulness <strong>of</strong> conjunctivalbiopsy as diagnostic technique. Arch Soc Esp Oftalmol76:31–3698. S<strong>and</strong>ers TE, Ackerman LV, Zimmerman LE (1962) Epi<strong>the</strong>lialtumors <strong>of</strong> <strong>the</strong> lacrimal gl<strong>and</strong>. A comparison <strong>of</strong> <strong>the</strong> pathologic<strong>and</strong> clinical behavior with those <strong>of</strong> <strong>the</strong> salivary gl<strong>and</strong>s. AmJ Surg 104:657–66599. Schuster V, Seregard S (2003) Ligneous conjunctivitis. SurvOphthalmol 48:369–388100. Sehgal VN, Ramesh V, Ghorpade A (1984) Naevus sebaceousassociated with ocular dermolipoma. J Dermatol 11:97–98101. Seitz B, Henke V (1995) Mucoepidermoid carcinoma <strong>of</strong> <strong>the</strong>epibulbar connective tissue with diffuse intraocular epi<strong>the</strong>lialinvasion. Klin Monatsbl Augenheilkd 207:264–265102. Sekundo W, Augustin AJ (2001) Differences in <strong>the</strong> composition<strong>of</strong> inflammatory cell infiltrate in lens-induced uveitisunder <strong>the</strong>rapy with allopurinol or steroids. Eur J Ophthalmol11:264–268103. Shields JA, Shields CL (2004) Orbital cysts <strong>of</strong> childhood –classification, clinical features, <strong>and</strong> management. Surv Ophthalmol49:281–299104. Shirai K, Saika S, Okada Y, Oda S, Ohnishi Y (2004) Histology<strong>and</strong> immunohistochemistry <strong>of</strong> fibrous posterior capsuleopacification in an infant. J Cataract Refract Surg 30:523–526105. Sjo NC, Heegaard S, Prause JU, von Buchwald C, LindebergH (2001) Human papillomavirus in conjunctival papilloma.Br J Ophthalmol 85:785–787106. Sjoegren H (1933) Angeborenes Fehlen der Tranensekretionund der Keratoconjunctivitis sicca bei Kindern. KlinMonatsbl Augenheilkd 22:554–559107. Sjoegren H (1933) Zur Kenntnis der Keratoconjunctivitis sicca(Keratitis filiformes bei hyp<strong>of</strong>unktion der Tranendrusen).Acta Opthalmol 2:1–151


310 M.R. Canninga-Van Dijk10108. Skotnitsky C, Sankaridurg PR, Sweeney DF, Holden BA(2002) General <strong>and</strong> local contact lens induced papillary conjunctivitis(CLPC). Clin Exp Optom 85:193–197109. Spencer WH, Fisher JJ (1959) The association <strong>of</strong> keratoconuswith atopic dermatitis. Am J Ophthalmol 47:332–344110. Spraul CW, Grossniklaus HE (1996) Tumors <strong>of</strong> <strong>the</strong> cornea<strong>and</strong> conjunctiva. Curr Opin Ophthalmol 7:28–34111. Spraul CW, Lang GK (1996) Oncocytoma <strong>of</strong> <strong>the</strong> conjunctiva.Klin Monatsbl Augenheilkd 209:176–177112. Starck T, Kenyon KR, Hanninen LA, Beyer-Machule C, FabianR, Gorn RA, McMullan FD, Baum J, McAdam KP (1991)Clinical <strong>and</strong> histopathologic studies <strong>of</strong> two families with latticecorneal dystrophy <strong>and</strong> familial systemic amyloidosis(Meretoja syndrome). Ophthalmology 98:1197–1206113. Stewart H, Black GC, Donnai D, Bonshek RE, McCarthy J,Morgan S, Dixon MJ, Ridgway AA (1999) A mutation withinexon 14 <strong>of</strong> <strong>the</strong> TGFBI (BIGH3) gene on chromosome 5q31causes an asymmetric, late-onset form <strong>of</strong> lattice corneal dystrophy.Ophthalmology 106:964–970114. Suchecki JK, Donshik P, Ehlers WH (2003) Contact lenscomplications. Ophthalmol Clin North Am 16:471–484115. Thorne JE, Jabs DA, Nikolskaia O, Anhalt G, Nousari HC(2002) Discoid lupus ery<strong>the</strong>matosus <strong>and</strong> cicatrizing conjunctivitis:clinicopathologic study <strong>of</strong> two cases. Ocul ImmunolInflamm 10:287–292116. Thorne JE, Jabs DA, Nikolskaia OV, Mimouni D, AnhaltGJ, Nousari HC (2003) Lichen planus <strong>and</strong> cicatrizing conjunctivitis:characterization <strong>of</strong> five cases. Am J Ophthalmol136:239–243117. To K, Adamian M, Dryja TP, Berson EL (2002) Histopathologicstudy <strong>of</strong> variation in severity <strong>of</strong> retinitis pigmentosa dueto <strong>the</strong> dominant rhodopsin mutation Pro23His. Am J Ophthalmol134:290–293118. Tranos L (1954) M<strong>and</strong>ibul<strong>of</strong>acial dysostosis associated withdermolipoma <strong>of</strong> <strong>the</strong> conjunctiva. Am J Ophthalmol 37:354–359119. Tuft SJ, Kemeny DM, Dart JK, Buckley RJ (1991) Clinical features<strong>of</strong> atopic keratoconjunctivitis. Ophthalmology 98:150–158120. Tunc M, Char DH, Crawford B, Miller T (1999) Intraepi<strong>the</strong>lial<strong>and</strong> invasive squamous cell carcinoma <strong>of</strong> <strong>the</strong> conjunctiva:analysis <strong>of</strong> 60 cases. Br J Ophthalmol 83:98–103121. Usui T, Sawaguchi S, Abe H, Iwata K, Oyanagi K (1993) Conjunctivalbiopsy in adult form galactosialidosis. Br J Ophthalmol77:165–167122. Uy HS, Pineda R II, Shore JW, Polcharoen W, Jakobiec FA,Foster CS (1999) Hypertrophic discoid lupus ery<strong>the</strong>matosus<strong>of</strong> <strong>the</strong> conjunctiva. Am J Ophthalmol 127:604–605123. Waring GO III, Rodrigues MM, Laibson PR (1978) Cornealdystrophies. I. Dystrophies <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium, Bowman’s layer<strong>and</strong> stroma. Surv Ophthalmol 23:71–122124. Weber CM, Eichenbaum JW (1997) Acute red eye. Differentiatingviral conjunctivitis from o<strong>the</strong>r, less common causes.Postgrad Med 101:185–196125. Wilhelmus KR, Robinson NM, Tredici LL, Jones DB (1986)Conjunctival cytology <strong>of</strong> adult chlamydial conjunctivitis.Arch Ophthalmol 104:691–693126. Wizigmann-Voos S, Plate KH (1996) <strong>Pathology</strong>, genetics<strong>and</strong> cell biology <strong>of</strong> hemangioblastomas. Histol Histopathol11:1049–1061127. Woerdt van der A (2000) Lens-induced uveitis. Vet Ophthalmol3:227–234128. Wolff SM (1972) The ocular manifestations <strong>of</strong> congenital rubella.Trans Am Ophthalmol Soc 70:577–614129. Woodward EG, Morris MT (1990) Joint hypermobility inkeratoconus. Ophthalmic Physiol Opt 10:360–362130. Wu SX (1987) Neur<strong>of</strong>ibromatosis accompanied by congenitalglaucoma (report <strong>of</strong> 2 cases). Yan Ke Xue Bao 3:43–46131. Yeung L, Tsao YP, Chen PY, Kuo TT, Lin KK, Lai LJ (2004)Combination <strong>of</strong> adult inclusion conjunctivitis <strong>and</strong> mucosaassociatedlymphoid tissue (MALT) lymphoma in a youngadult. Cornea 23:71–75132. Zaidman GW (1997) The ocular manifestations <strong>of</strong> Lyme disease.Int Ophthalmol Clin 37:13–28133. Zhang J (1991) Histo- <strong>and</strong> ultrahistopathology <strong>of</strong> <strong>the</strong> anteriorchamber angle in congenital glaucoma. Zhonghua Yan KeZa Zhi 27:151–153134. Zhu JB, Li B, Sun XL, Li LQ, Shi JT, An YZ (2004) Clinical<strong>and</strong> pathological features <strong>of</strong> 273 cases <strong>of</strong> lacrimal epi<strong>the</strong>lialtumors. Zhonghua Yan Ke Za Zhi 40:220–224


Subject IndexAabscess 184peritonsillar 184abuse <strong>of</strong> <strong>the</strong> voice 208accessory tragus 240external ear 240acinic cell carcinoma 60sinonasal 60Addison disease 88adenocarcinoma 149, 150, 151, 182,222, 242basal cell 151ceruminal gl<strong>and</strong>s 242cribiform <strong>of</strong> <strong>the</strong> tongue 150larynx 222nasopharynx 182polymorphous low-grade 149adenoid cystic carcinoma 59, 182, 222,242ceruminal gl<strong>and</strong>s 242larynx 222nasopharynx 182sinonasal 60adenoid squamous cell carcinoma 22adenoma 48, 137, 140, 141, 143, 144, 158,174, 214, 240, 241, 247, 248basal cell 141canalicular 143ceruminal gl<strong>and</strong>s 240ceruminal gl<strong>and</strong>s pleomorphic 241congenital pleomorphic 140larynx pleomorphic 214middle ear 247middle ear neuroendocrine 248middle ear plasmacytoid 248nasopharyngeal pituitary 174pleomorphic 48, 137pleomorphic metastasising 158sebaceous 144sinonasal pituitary 48adenomatoid hyperplasia 134salivary gl<strong>and</strong>s 134adenomatoid odontogenic tumour 112adenosquamous carcinoma 23differential diagnosis 23agenesis 132salivary gl<strong>and</strong>s 132aggressive papillary tumour 248middle ear 248amalgam tattoo 86ameloblastic carcinoma 119ameloblastic fibro-dentinoma 116ameloblastic fibro-odontoma 116ameloblastic fibro-odontosarcoma 120ameloblastic fibrodentinosarcoma 120ameloblastic fibroma 116granular cell 116ameloblastic fibrosarcoma 120ameloblastoma 110, 111, 112, 119acanthomatous 111basaloid 111basal cell 111desmoplastic 111granular cell type 111malignant 119metastasing 119peripheral 112unicystic 111amyloidosis 43, 191, 211, 305larynx 211sinonasal 43Waldeyer‘s ring 191aneurysmal bone cyst 124angi<strong>of</strong>ibroma 50, 175nasopharyngeal 50, 175angiolymphoid hyperplasiawith eosinophilia 239external ear 239aniridia 294with Wilm‘s tumour 294aphthae 77herpetiform 77major 77minor 77aplasia 132salivary gl<strong>and</strong>s 132aspergillosis 44, 203invasive 44larynx 203sinonasal 44astrocytic hamartoma 301in tuberous sclerosis 301<strong>of</strong> retina 301atresia 132salivary gl<strong>and</strong>s 132atypical hyperplasia 10BB-cell lymphoma 61sinonasal 61basaloid squamous cell carcinoma 20basal <strong>and</strong> parabasal cell hyperplasia 9basal cell carcinoma 242external ear 242Behçet disease 78Bell‘s palsy 255blastomycosislarynx 203Bohn’s nodules 109botryoid odontogenic cyst 107branchial 265arch 265cleft 265cyst 265fistula 265pouch 265sinus 265branchial cleft cyst 238first 238branchiogenic carcinoma 267Ccalcifying cystic odontogenic tumour 118calcifying epi<strong>the</strong>lial odontogenic tumour112calcifying odontogenic cyst 118c<strong>and</strong>idiasis 203larynx 203c<strong>and</strong>idosis 81, 82chronic hyperplastic 82carcinoma 119, 144, 146, 147, 150, 151,152, 154, 156, 157, 158, 159, 180, 189,252, 267, 290acinic cell 144adenoid cystic 147branchiogenic 189, 267epi<strong>the</strong>lial-myoepi<strong>the</strong>lial 150ex pleomorphic adenoma 156ex pleomorphic adenoma,non-invasive 157hyalinising clear cell 151lymphoepi<strong>the</strong>lial 158middle ear metastatic 252mucoepidermoid 146myoepi<strong>the</strong>lial 152nasopharyngeal keratinising 180nasopharyngeal non-keratinising 180nasopharyngeal undifferentiated 180oncocytic 156primary intraosseous 119salivary duct 154sebaceous 158sebaceous <strong>of</strong> conjunctiva 290small cell 159carcinoma in situ 11carcinosarcoma 157salivary gl<strong>and</strong> 157


312 Subject Indexcataract 297cellulitis 306in orbit 306cemento-ossifying fibroma 121cementoblastoma 115cementoma 124familial gigantiform 124ceruminoma 240ear canal 240chalazia 304cheilitis granulomatosa 92cherubism 124Chickenpox 73Chievitz’s organ 72chlamydia trachomatis 287cholesteatoma 239, 244, 246, 259cerebellopontine angle 259congenital 246external canal 239middle ear 244cholesterol granuloma 43, 259maxillary sinus 43petrous apex 259chondrodermatitis nodularis helicis 239chondroid chordoma 125chondromas 217larynx 217chondrosarcoma 125, 183, 223larynx 223nasopharynx 183chordoma 125, 183chondroid 125nasopharynx 183choristoma 247glial middle ear 247salivary gl<strong>and</strong> middle ear 247sebaceous middle ear 247clear cell odontogenic carcinoma 120clear cell odontogenic tumour 120cocaine abuse 46nasal 46coccidioidomycosislarynx 203complex odontoma 117compound odontoma 117condyloma acuminatum 2conjunctivitis 286, 287, 288acute 286chronic 287inclusion 287, 288in dermatologic diseases 288ligneous 287contact ulcer 210glottic 210corneal 294failed graft 294corneal dystrophies 293Avellino 293Cogan‘s 293Fuchs 293granular 293lattice 293macular 293Meesman‘s 293Reis-Buckler‘s 293corneal ulceration 292in systemic disease 292coxsackie virus 73craniopharyngioma 118, 174nasopharynx 174Crohn’s disease 91cryptococcosis 203larynx 203cylindrical cell carcinoma 52sinonasal 52cyst 42, 105, 106, 107, 108, 109, 124, 135,173, 174, 179, 199, 200, 268, 269, 270,271, 273, 285, 303, 306aneurysmal bone 124benign lymphoepi<strong>the</strong>lial 136botryoid odontogenic 107bronchial 270bronchogenic 270calcifying odontogenic 118conjunctival inclusion 285dentigerous 106dermoid 271, 303epidermal 303eruption 106gingival 108gl<strong>and</strong>ular odontogenic 107gl<strong>and</strong>ular retention 179larynx ductal 199larynx epidermoid 200larynx oncocytic 200larynx saccular 199lateral periodontal 107lymphoepi<strong>the</strong>lial 273nasolabial 109nasopalatine duct 109nasopharyngeal branchial 173nose dermoid 42<strong>of</strong> orbit 306paradental 106parathyroid 270primordial 107radicular 105Rathke’s cleft 174residual 105salivary duct 135salivary gl<strong>and</strong>s 135sialo-odontogenic 107simple bone 109solitary bone 109surgical ciliated 109thymic 269thyroglossal duct 268Tornwaldt’s 173traumatic bone 109cystadenoma 144mucinous 144oncocytic 144papillary 144cystic hygroma 272cystic lymphoid hyperplasia <strong>of</strong> AIDS 136cytomegalovirus 133, 254cochlear infection 254salivary gl<strong>and</strong>s 133Ddacryoadenitis 302dental follicle 114, 115, 117dental papilla 114dentigerous cyst 106dentinogenic ghost cell tumour 118dermatitis herpetiformis 76dermoid 285dermolipoma 285desmoid fibromatosis 49sinonasal 49desmoplastic ameloblastoma 111desmoplastic fibroma 115diph<strong>the</strong>ria 202larynx 202dysplasia 5, 8Eendodermal sinus tumour 161endolymphatic sac tumour 259temporal bone 259endophthalmitis 295eosinophilic angiocentric fibrosis 43sinonasal 43eosinophilic ulcer 79epidermoid cyst 200epiglottitis 202acute 202Epstein-Barr virus 133salivary gl<strong>and</strong>s 133Epstein’s pearls 109epulis 90, 93fibrous 90giant cell 93eruption cyst 106ery<strong>the</strong>ma multiforme 77erythroplakia 6exostosis 241ear canal 241Ffacial paralysis 255familial gigantiform cementoma 124fibro-osseous lesions 121fibrosarcoma 62sinonasal 62fibrous dysplasia 121fibrous histiocytoma 49sinonasal 49focal osseous dysplasia 124focal epi<strong>the</strong>lial hyperplasia 2Fordyce granules 72foreign body 210frictional keratosis 86Ggenetic alterations 161salivary gl<strong>and</strong> tumours 161


Subject Index 313geographic tongue 85giant-cell hyalin angiopathy 93giant cell epulis 93, 124giant cell fibroma 94giant cell granuloma 124central 124peripheral 93, 124gingival 91fibromatosis 91hypertrophy 91gingival cyst 108gingival hyperplasia 93inflammatory 93gingivitis 79, 82, 92acute necrotising 79chronic marginal 92desquamative 82gl<strong>and</strong>ular odontogenic cyst 107glaucoma 294, 296neur<strong>of</strong>ibromatosis 294Sturge-Weber syndrome 294glial heterotopia 43glioma 301, 302in neur<strong>of</strong>ibromatosis 302nasal 43<strong>of</strong> optic nerve 302<strong>of</strong> optic pathway 301gout 206, 238external ear 238larynx 206granular cell ameloblastic fibroma 116granular cell odontogenic fibroma 116granular cell tumour 96, 216larynx 216granuloma 206, 210, 211glottic 210glottic intubation 210vocal cord teflon 206granulomatous infections 133salivary gl<strong>and</strong>s 133granuloma annulare 304Graves‘ disease 305Hhaemangioblastoma 301<strong>of</strong> retina 301haemangioma 48, 95, 214, 215, 272, 301<strong>of</strong> choroid 301sinonasal 48larynx 214subglottic 215haemangiopericytoma 48sinonasal 48hairy tongue 85hamartoma 42, 178, 247middle ear 247nasopharynx 178respiratory epi<strong>the</strong>lial adenomatoid 42,178sinonasal 42h<strong>and</strong>-foot-<strong>and</strong>-mouth disease 73herpangina 74herpes simplex 72Hertwig sheath <strong>of</strong> 104heterotopic brain 174nasopharyngeal 174hidrocystoma 303Hippel-Lindau disease 248middle ear 248histoplasmosis 203larynx 203hygroma 95, 272cystic 95, 272hyperacidic granulomas 211hyperplastic vitreous 294hypoplasia 132salivary gl<strong>and</strong>s 132Iidiopathic pseudocystic chondromalacia238pinna 238inflammatory my<strong>of</strong>ibroblastic tumour213larynx 213intercalated duct hyperplasia 151intestinal-type adenocarcinoma 58sinonasal 58intraepi<strong>the</strong>lial neoplasia 289<strong>of</strong> conjunctiva 289Kkeloid 240ear 240keratinising cysts 200keratin granuloma 239external ear 239keratitis 292herpes simplex 292keratoameloblastoma 111keratoconjunctivitis sicca 288keratoconus 292syndrome-associated 292keratocyst 107keratocystic odontogenic tumour 108keratocystoma 137keratosis 5keratosis obturans 239ear canal 239Kimura‘s disease 239external ear 239Küttner tumour 133Llabyrinthitis 255lacrimal gl<strong>and</strong> 303tumours 303laryngocele 199, 271laryngotracheobronchitis 202lateral periodontal cyst 107Laugier Hunziker syndrome 89leiomyoma 49sinonasal 49leiomyosarcoma 63sinonasal 63leishmaniasis 45, 203larynx 203nasal 45leprosy 45, 203larynx 203nasal 45leukoplakia 5, 6, 85hairy 85lichenoid drug eruption 83lichen planus 82linear IgA disease 76lipoma 258inner ear 258Lipschutz bodies 72Ljubljana grading system 9low-grade adenocarcinoma 60sinonasal 60lupus ery<strong>the</strong>matosus 83lymphadenoma 144sebaceous 144lymphangioma 95, 272lymphoepi<strong>the</strong>lial carcinoma 24differential diagnosis 25larynx 24nasopharynx 24oropharynx 24lymphoepi<strong>the</strong>lial sialadenitis 163lymphoid hyperplasia 184palatine tonsil 184pharyngeal tonsil 184lymphoma 61, 164, 189, 190, 224larynx 224MALT 164sinonasal 61Waldeyer‘s ring 189Waldeyer‘s ring Hodgkin 190Waldeyer‘s ring MALT 190Waldeyer‘s ring mantel cell 189Waldeyer‘s ring NK/T-cell 190lymph nodes 264cervical groups 264Mmacule 86oral melanotic 86Malassez rests <strong>of</strong> 104malignant epi<strong>the</strong>lial odontogenic ghostcell tumour 120malignant fibrous histiocytoma 62sinonasal 62malignant melanoma 243external ear 243malignant peripheral nerve sheath tumour63sinonasal 63Marfan’s syndrome 107melanoacanthoma 87melanoma 55, 87, 225, 291, 299in situ 88larynx 225<strong>of</strong> conjunctiva 291


314 Subject Index<strong>of</strong> uveal tract 299sinonasal 55melanosis 89, 291<strong>of</strong> conjunctiva 291smoker’s 89melanotic neuroectodermal tumour <strong>of</strong>infancy 126melanotic nevi 243external ear 243Melkersson Rosenthal syndrome 92Ménière‘s disease 256meningioma 50, 123, 125, 179, 251,258, 302chordoid 125middle ear 251nasopharynx 179sinonasal 50inner ear 258metastases 160, 225, 307larynx 225<strong>of</strong> orbit 307salivary gl<strong>and</strong>s 160metastatic renal cell carcinoma 61sinonasal 61metastatic tumour 191, 302<strong>of</strong> <strong>the</strong> eye 302nasopharynx 191Miescher’s syndrome 92migratory stomatitis 85Mikulicz syndrome 302mucocele 43, 132, 135extravasation 132sinonasal 43mucoepidermoid carcinoma 61, 222larynx 222sinonasal 61mucormycosissinonasal 44multiple endocrine neoplasia syndrome96mumps 133salivary gl<strong>and</strong>s 133muscular dystrophy 207oculopharyngeal 207myoepi<strong>the</strong>lioma 140, 152malignant 152myospherulosis 43sinonasal 43myxoma 114Nnaevi 87blue 87intramucosal 87melanocytic 87naevus 84, 290, 298<strong>of</strong> conjunctiva 290<strong>of</strong> uveal tract 298oral epi<strong>the</strong>lial 84unius lateris 84white sponge 84nasolabial cyst 109nasopalatine duct cyst 109nasopharyngeal-type undifferentiatedcarcinomas 54sinonasal 54neck triangles 264neck dissection 278examination 279level 278types 279necrobiotic xanthogranuloma 305necrotising sialometaplasia 133larynx 211neurilemmoma 96neurinoma 96neuritis 302optic 302neuroendocrine carcinoma 220, 221larynx 220larynx moderately differentiated 220larynx poorly differentiated 221larynx well differentiated 220neur<strong>of</strong>ibroma 49, 96sinonasal 49neur<strong>of</strong>ibromatosis 96, 124, 258acoustic 258neuromas 96mucosal 96nevoid basal cell carcinoma syndrome 107NK/T cell lymphoma 61sinonasal 61nodule 208, 211glottic metaplastic elastic cartilaginous211vocal cord 208Noonan’s syndrome 124Oochronosis 238external ear 238odonto-ameloblastoma 118odontogenic carcinosarcoma 121odontogenic epi<strong>the</strong>lial hamartoma 115odontogenic fibroma 114, 115, 116granular cell 115, 116peripheral 115odontogenic ghost cell carcinoma 120odontogenic ghost cell tumour 120malignant epi<strong>the</strong>lial 120odontogenic gingival epi<strong>the</strong>lial hamartoma112odontogenic granular cell tumour 115odontogenic keratocyst 107odontogenic myxoma 114odontoma 117complex 117compound 117oedema 207, 208larynx angioneurotic 207vocal cord Reinke‘s 208olfactory neuroblastoma 57oncocytic hyperplasia 135oncocytic metaplasia 134, 200oncocytoma 143, 214, 289larynx 214<strong>of</strong> conjunctiva 289oncocytosis 134ophthalmitis 296sarcoidosis 295sympa<strong>the</strong>tic 296optic atrophy 302or<strong>of</strong>acial granulomatosis 92osseous dysplasia 123, 124florid 124focal 124periapical 124ossifying fibroma 90, 115, 121, 122, 241juvenile psammomatoid 122juvenile trabecular 122peripheral 90, 115temporal bone 241osteoblastoma 115osteogenesis imperfecta 254stapes 254osteoma 125, 241ear canal 241Gardner’s syndrome 125osteomyelitis 104osteopetrosis 254otic 254osteoporotic bone marrow defect 109osteosarcoma 183nasopharynx 183otitis 237, 244malignant externa 237media 244otosclerosis 252bony labyrinth 252stapes footplate 252PPaget‘s disease 253, 256inner ear 256temporal bone 253panophthalmitis 295papillary adenocarcinoma 182nasopharynx 182papillary hyperplasia 90papillary squamous cell carcinoma 19differential diagnosis 20papilloedema 302papilloma 46, 47, 144, 178, 249, 288ductal 144exophytic 46middle ear schneiderian 249nasopharynx inverted 178<strong>of</strong> conjunctiva 288sinonasal 46sinonasal exophytic 46sinonasal inverted 46sinonasal oncocytic 47papillomatosis 3recurrent respiratory 3paracoccidioidomycosis 203larynx 203paradental cyst 106paraganglioma 50, 215, 249, 273middle ear jugulotympanic 249multicentric jugulotympanic 249sinonasal 50


Subject Index 315solitary jugulotympanic 249larynx 215pemphigoid 75cicatricial 75mucous membrane 75pemphigus 74paraneoplastic 75vegetans 74vulgaris 74periapical osseous dysplasia 124perichondritis 237pinna 237periostitis 104peripheral ameloblastoma 112petrositis 255bacterial 255Peutz Jeghers syndrome 89phonotrauma 209phtisis bulbi 298pigmentation 89, 90drugs associated 90heavy metals 90racial 89Pindborg tumour 112pinguecula 286plasmacytoma 62, 190, 224larynx 224sinonasal 62Waldeyer‘s ring 190polycystic dysgenetic disease 135polymorphous low-grade adenocarcinoma182nasopharynx 182polyp 41, 90, 175, 184, 187, 208allergic sinonasal 41antrochoanal 41antronasal 42atypical fibroblasts 41fibroepi<strong>the</strong>lial 90immobile cilia syndrome 41Kartagener’s syndrome 41mucoviscidosis 41nasopharynx hairy 175oropharynx hairy 175palatine tonsil hairy 184tonsillar lymphangiomatous 187vocal cord 208postintubation granuloma 211presbyacusis 260primary intraosseous carcinoma 119primitive neuroectodermal tumour 58sinonasal 58primordial cyst 107proliferative verrucous hyperplasia 6pseudotumour 306<strong>of</strong> orbit 306psoriasis 86pterygium 286pulse granuloma 93pyogenic granuloma 93Rradicular cyst 105ranula 132, 271Reinke’s oedema 208Reinke’s space 208Reiter disease 78relapsing polychondritis 205, 238larynx 205pinna 238residual cyst 105retention cyst 200retinal detachment 298retinal dysplasia 294retinal ischaemia 298retinitis pigmentosa 298retinoblastoma 300retinochoroiditis 295toxoplasma gondii 295retrolental fibroplasia 294rhabdomyosarcoma 63, 183, 252middle ear 252nasopharynx 183sinonasal 63rheumatoid arthritis 204larynx 204rhinitis 40allergic 40atrophic 41bacterial 40hypertrophic 41viral 40rhinoscleroma 45, 203larynx 203nasal 45rhinosinusitis 40bacterial 40rhinosporidiosis 44sinonasal 44rhomboid glossitis 78Riga-Fede disease 79Rubella syndrome 294congenital 294Rushton bodies 105, 108Ssaccular cysts 199salivary duct carcinomas 60sinonasal 60salivary gl<strong>and</strong> anlage tumour 140, 175nasopharynx 175salivary gl<strong>and</strong> neoplasms 273cervical cystic 273sarcoidosis 45, 191, 204larynx 204sinonasal 45Waldeyer‘s ring 191sarcoma 183, 224larynx 224larynx granulocytic 224larynx mast cell 224nasopharynx granulocytic 183schwannoma 49, 96, 257acoustic 257sinonasal 49sclerosing polycystic sialadenopathy 136Serres rests <strong>of</strong> 104sialadenosis 133sialo-odontogenic cyst 107sialoblastoma 161simple bone cyst 109sinusitis 44allergic mucinous 44sinus histiocytosis with massive lymphadenopathy212larynx 212Sjögren‘s syndrome 288small cell neuroendocrine carcinoma 54sinonasal 54smoker’s keratosis 84solitary bone cyst 109solitary fibrous tumour 48, 179nasopharynx 48, 179sinonasal 48spindle cell carcinoma 16differential diagnosis 17, 21squamous cell carcinoma 13, 15, 25, 51,96, 187, 218, 243, 250acantholytic 22adenoid 22alcohol 14basaloid 20conventional 13cystic metastasis 187differential diagnosis 15external ear 243glottic 218grading 15hypopharyngeal 219invasive front 15larynx aetiology 218larynx epidemiology 218metastases 25microinvasion 13micrometastasis 27middle ear 250molecular pathology 28nasal vestibule 51pseudogl<strong>and</strong>ular 22second primary tumours 25sinonasal 51smoking 14spindle 16stromal reaction 15subglottic 219supraglottic 218transglottic 219treatment <strong>and</strong> prognosis 15Waldeyer‘s ring 187squamous cell hyperplasia 9squamous cell papilloma 2, 187, 214larynx 214malignant transformation 4oral cavity 2oropharyngeal 187oropharynx 2tonsillar 187nasal vestibule 46squamous intraepi<strong>the</strong>lial lesions 4, 217genetic progression 12larynx 5, 12, 217oral cavity 5oropharynx 5


316 Subject Indexsquamous odontogenic tumour 113steroid injections 46nasal 46stomatitis nicotina 84subglottic stenosis 206idiopathic 206surgical ciliated cyst 109syringocystadenoma papilliferum 241ceruminal gl<strong>and</strong>s 241TTangier’s disease 190Waldeyer‘s ring 190teratocarcinosarcoma 63sinonasal 63teratoma 175, 272cervical 272nasopharynx congenital 175thymoma 273cervical 273thyroid 94, 201, 268ectopic 268ectopic lingual 94larynx aberrant 201tonsillitis 184, 185bacterial suppurative 184viral 185tori 125tracheopathia osteochondroplastica 202trachoma 287stages 287traumatic bone cyst 109trichinosis 203larynx 203tuberculosis 45, 81, 133, 191, 202, 247larynx 202middle ear 247nasal 45salivary gl<strong>and</strong>s 133Waldeyer‘s ring 191tuberous sclerosis 115tubulopapillary carcinoma 61sinonasal 61Turner‘s syndrome 95, 272tympanosclerosis 244Tzanck cells 74Uundifferentiated carcinoma 53sinonasal 53unicystic ameloblastoma 111unknown primary tumour 274uveitis 296lens-induced 296Vvaricella zoster 73verruca vulgaris 2verrucous carcinoma 17differential diagnosis 18verrucous hyperplasia 7vertigo 256positional 256vocal cord polyp 209voice changes 209voice re-education 211voice rehabilitation 208, 210voice rest 211von Hippel-Lindau disease 301von Recklinghausen disease 96WWarthin’s tumour 142Wegener‘s disease 45, 80, 203larynx 203sinonasal 45Xxan<strong>the</strong>lasmata 305xanthoma 95verruciform 95ZZenker‘s diverticulum 201hypopharynx 201

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!