Academia.eduAcademia.edu
8 Immunomodulators HESHAM EL ENSHASY1,2 CONTENTS I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . II. Immunosuppressants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . A. Cyclosporins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1. Chemistry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2. Biosynthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3. Production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4. New Generations . . . . . . . . . . . . . . . . . . . . . . . . . . . B. Mycophenolic Acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1. Chemistry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2. Biosynthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3. Production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . C. Mizoribine. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . D. Other Immunosuppressants. . . . . . . . . . . . . . . . . . . 1. Ovalicins and Fumagillins . . . . . . . . . . . . . . . . . 2. Gliotoxin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3. Trichopolyns. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4. Myriocin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5. Flavidulols . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6. Kobiins. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7. Mycestericins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8. Terprenins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9. FR901483. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10. Colutellin A. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . III. Mushroom Immunomodulators . . . . . . . . . . . . . . . . . . A. Lectins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . B. Terpenoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . C. Polysaccharides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . D. Fungal Immunomodulator Proteins . . . . . . . . . . E. Industrial Production of Mushroom Immunomodulators. . . . . . . . . . . . . . . . . . . . . . . . . . . . IV. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . I. Introduction 165 166 166 168 168 170 171 171 173 174 174 175 176 176 176 178 179 179 179 180 180 180 180 180 181 181 181 184 184 185 185 1 Chemical Engineering Pilot Plant (CEPP), Faculty of Chemical and Natural Resources Engineering, Universiti Teknologi Malaysia (UTM), 81310 Skudai, Johor, Malaysia; e-mail: hesham@ utm.my 2 Bioprocess Development Department, Mubarak City for Scientific Research and Technology Applications (MuCSAT), New Burg Al Arab, Alexandria, Egypt Immunomodulators are actually natural products of the immune system (Pirofski and Casadevall 2006). The immune system of healthy organism produces a diverse range of metabolites to keep the body in homeostasis condition. An immunomodulator may be defined as a substance, of biological or synthetic origin, which can stimulate, suppress or modulate any of the components of the immune system including both innate and adaptive arms of the immune response (Agawal and Singh 1999). In clinical practice, immunomodulators are classified into three main categories: 1. Immunosuppressants are agents that somehow inhibit the immune system. They can be used for the control of pathological immune response after organ transplantation and for the treatment of autoimmune diseases, hypersensitivity immune reactions as well as immune pathology associated with infections. 2. Immunostimulators are agents that stimulate the immune system by inducing the activation or increasing activity of any of its components. They enhance the body’s resistance against allergy, infection, cancer and autoimmunity. 3. Immunoadjuvants are agents used to enhance the vaccine efficacy. This can be also considered as a specific immune stimulator effect. The industrial importance of immunomodulators is based on their large market value. The market size of immunomodulators was evaluated at US $43 billion in 2006, and is expected to grow at a compound annual growth rate (CAGR) of 13% to reach US $80 billion by 2011 (Research and Market 2007). Although some potential immunomodulating substances can be chemically synthesized and have been successfully tested for modulation of the immune system [e.g. synthetic muramyl dipeptide (MDP) analogues], research activities Industrial Applications, 2nd Edition The Mycota X M. Hofrichter (Ed.) © Springer-Verlag Berlin Heidelberg 2010 166 Hesham El Enshasy in this field have been focused on immunomodulatory active compounds from natural resources. Among them, fungal immunomodulators represent the most interesting group of metabolites. This review outlines the current state of knowledge on fungal immunomodulators used already for different medical applications and discusses the future potential of new compounds of fungal origin. II. Immunosuppressants The importance of immunosuppressants arose in the mid of the twentieth century in the context of new developments in organ transplantation. The principal goal of immunosuppressant application in organ transplantation is to minimize the risk of allo-graft rejection or graft-dysfunction by achieving adequate immunosuppression, yet also to ensure that the level of immunosuppression does not contribute to long term morbidity (Patel and Kobashigawa 2008). The first organ transplantation was performed in 1933 when a kidney was transplanted from a cadaver. Total lymphoid irradiation was used for the immune suppression but the tissue was rejected and the patient eventually died. This was followed by the use of corticosteroids as immunosuppressive agents, but unfortunately, these steroids as such did also not give the positive results expected. In the early 1960s, cytotoxic agents such as modified corticosteroids were introduced to suppress the immune system after organ transplantation (Khan 2008). The first clinically used fungal immunosuppressive agent was introduced into the market in the mid of 1980s when cyclosporine became available for clinical applications after getting its approval from the United States Food and Drug Administration (US FDA). This was one of the most important milestones in the history of organ transplantation. Beside their important roles in organ transplantation, immunosuppressive agents are used for other applications, for example, in the prevention of the newborn Rh hemolytic disease (Contreas and DeSilva 1994) or for the treatment of some autoimmune diseases. The chemical structures of the three main fungal immunosuppressive agents (cyclosporine, mycophenolic acid, mizoribine), which all are used in organ transplantation and for other medical applications, are shown in Fig. 8.1. A. Cyclosporins Cyclosporins (Cys) are a family of neutral, high lipophilic, cyclic undecapeptides containing some unusual amino acids and having a remarkable spectrum of biological activities. The first member of this class of compounds was named cyclosporine A. To date, more than 30 members of this family of compounds have been isolated from natural resources and were classified as cyclosporins A to Z (CyA–Z; Traber et al. 1982, 1987). CyA was originally described as an antifungal peptide with a narrow spectrum of efficacy. However, the interest in this compound only increased significantly after the demonstration of its specific immunosuppressive activity. In 1983, Sandoz first introduced a cyclosporine-A-based drug, Sandimmun, into the market. The modified form of this drug with increased bioavailability, Neoral, became available on the market in 1994 in form of soft gelatins and oral applicable solutions. Since then, Sandimmun and Neoral have been Novartis’ leading pharmaceutical products and these drugs generated a revenue of US $1.216 billion in 1997 (Svarstad et al. 2000). Nowadays, based on market research data, only five members of the cyclosporin family, namely CyA (CAS 59865-13-3), CyB (CAS 63775-95-1), CyC (CAS 59787-61-0), CyD (CAS 63775-96-2) and CyH (CAS 83602-39-5) are commercially available for pharmaceutical applications as immunosuppressive agents. According to their immunosuppressive mode of action, cyclosporins belong to the specific calcineurin inhibitors. Immunosuppressant activity is mediated through blocking the activation and proliferation of CD4þ- and CD8þ-T lymphocytes by inhibiting IL-2 production (Siekierka et al. 1989; Shibasaki et al. 2002). Under normal conditions, the binding of major histo-compatibility peptides to the T-cell receptors results in the formation of an activated form of calcium/calmodulin-dependent serine/threonine phosphatase calcineurin. This leads to dephosphorylation and nuclear translocation of the nuclear factor of activated T-cells (NF-AT), Subsequently, NF-AT binds genes encoding pro-inflamatory cytokine IL-2, resulting in an up-regulated gene transcription (Schreiber and Crabtree 1992; Butch 2008). CyA freely crosses lymphocyte membranes and forms complexes with the specific cytoplasmatic binding protein immunophilin cyclophilin A. The CyA-cyclophilin A complex inhibits calcineurin CH3 H3C CH3 CH3 CH3 O N O CH3 CH3 H3C N O CH3 NH2 OH N 2-Mycophenolic acid (MPA) C H3 CH3 H3CO O CH CH3 O O N O CH3 CH3 C O H3 H N N H C H3 N N O HO CH3 H N H3C O HO CH3 CH3 CH3 OH N O O N CH3 O OH OH O Immunomodulators H3C CH3 H3C N H O O O N N H3C CH3 O OH HO O O O H3CO CH3 1- cyclosporins 3- Mycophenolic acid 4- Mirozoribine Fig. 8.1. Chemical structure of the main clinically important fungal immunosuppressives compounds 167 168 Hesham El Enshasy activity and the nuclear translocation of NF-AT. This leads to the down-regulation of the proinflammatory molecules gene transcription and subsequently halts the production of IL-2 and TNF-a (Jorgensen et al. 2003). Currently, CyA is approved and used worldwide as an immunosuppressive drug to prolong organ and patient survival after kidney, liver, heart and bone-marrow transplants. CyA is available on the market under the trade name Sadimmune for both oral and intravenous applications. A nano-sized pre-concentrate formulation of CyA (CyA-MEPC or Neoral) exhibiting a better absorption characteristic is used orally in form of solutions or soft-gelatin capsules (Vonderscher and Meinzer 1994; Uchida et al. 2004). Besides the products of the market leader Novartis, several generic formulations are nowadays available and are often referred to as modified CyAs (Alloway 1999). 1. Chemistry CyA, the most important member of the cyclosporin family, is a cyclic undecapeptide with a 33membered ring composed of 11 lipophilic aliphatic amino acids, of which four are leucine and three are the non-proteogenic amino acids D-alanine, (4R)-4-[(E)-2-butenul]-4-methyl-L-threonine (Bmt) and L-a-aminobutyric acid. The full chemical name of CyA is 32-ethyl-2-[(E,1R,2R)-1-hydroxy2-methylhex-4-enyl]-3,6,9,12,14,17,21,27,30-nonamethyl-8,11,20,26-tetrakis-(2-methylpropyl)-5, 23-di-(propan-2-yl)-3,6,9,12,15,18,21,24,27,30,33undecaza-cyclotritriacontane-1,4,7,10,13,16,19,22, 25,28,31-undecone. The molecular formula and molecular weight of CyA are C62H111N11O12 and 1202.61 g mol–1, respectively. The non-proteogenic amino acids are found at the positions 1 (Bmt), 2 (L-a-amino butyric acid) and 8 (D-alanine). Remarkably, seven of the 11 peptide bonds are N-methylated, which has several important implications. First, the N-methylated peptide bonds and the cyclic structure of the molecule renders cyclosprins stable toward mammalian digestive and systemic proteases. Cyclosporin metabolism in animals and humans is exclusively carried out by cytochrome P450 enzymes catalyzing its oxidative transformation. Therefore, cyclosporins are not only well absorbed when given orally but also characterized by high and long-lasting plasma levels. A second consequence of the N-methylation pattern is rigid conformation in the non-polar environment characterized by intramolecular hydrogen bonds being oriented towards the hydrophobic environment (Kallen et al. 1997). Plain cyclosporin is difficult to crystallize on its own and therefore, was initially analyzed as crystalline iodo-cyclosporin. The structural analysis of such CyA crystals by X-ray diffraction revealed a rigid conformation (Loosli et al. 1985). The rigidity can be attributed to a number of unique structural properties. Predominantly, the four intra-molecular hydrogen bonds maintain it by stabilizing the backbone structure. Not least, this is evident from the increase in the number of backbone conformations observed in polar solvents due to the formation of inter-molecular hydrogen bonds with the solvent molecules (Kratochvil et al. 1999). In addition to the four intra-molecular hydrogen bonds, CyA exhibits a cis-amide bond between the N-methylleucine residues at positions 9 and 10. Moreover, the N-methyl moiety of MeVal in the loop makes backbone contacts, which further contribute to the rigidity of the structure (Velkov and Lawen 2003). 2. Biosynthesis The biosynthesis of bioactive peptides like cyclosporins proceeds non-ribosomally and is catalyzed by complex multi-functional enzymes termed non-ribosomal peptide synthetases (NRPS). Cyclosporin synthetase (CySyn) is one of the best studied enzyme complexes of this type and capable of catalyzing a total of at least 39 different reaction steps in the synthesis of cycloundecapaptides via an assembly belt-like mechanism: 11 amino acyladenylation reactions, ten transpeptidations, seven N-methylations, ten chain elongation reactions and a final cyclization reaction (Dittmann et al. 1994; Velkov and Lawen 2003). The enzyme consists of 11 protein modules, each being responsible for the recognition, activation and modification of one substrate (Lawen and Zocher 1990; Weber et al. 1994) and a small 12th module putatively responsible for cyclization. Based on the gene sequence and the established models for non-ribosomal peptide synthetases (Marahiel et al. 1997), each module of CySyn essentially consists of a central adenylation domain (A-domain; Immunomodulators 169 well. The biosynthesis of Bmt is catalyzed by a polyketide synthase (PKS) that forms the polyketide backbone by the head-to-tail condensation of four acetate units, resulting in a 3(R)-hydroxy-4-(R)-methyl-6-(E)-octenoic acid thioester; the C-methyl in the carbon chain is derived from AdoMet (Offenzeller et al. 1993). The polyketide 3(R)-hydroxy-4(R)-methyl-6-(E)-octenoyl-CoA is then transformed into the b-amino acid form which is utilized by CySyn as a substrate for cyclosporine biosynthesis. D-Alanine is provided by a distinct pyridoxal phosphate dependent alanine racemase (Hoffmann et al. 1994). The remaining amino acid constituents of the CyA molecule are synthesized by classic biosynthetic pathways, as confirmed by Senn et al. (1991) using 13C-labeling experiments. recognition, activation), a thiolation domain (T-domain, covalent binding of adenylated amino acid on phosphopantethein) and a condensation domain (C-domain; elongation step). During elongation, the activated amino acids are linked by peptide bonds leading to enzyme-bound nascent peptide chains. CySyn substrates include L-valine, L-leucine, a-amino butyric acid (Abu), 4-methylthreonine, and D-alanine. With the adenulation domain, cyclosporine synthetase generates the acyl-adenylated amino acids and then covalently binds the amino acid to phosphopantetheine through a thioester linkage. Seven of the substrate amino acids become N-methylated by S-adenosylmethionine via respective methyltransferase activites of CySyn. The final cyclization step releases CyA from the enzyme complex (Hoppert et al. 2001). L-alanine, L-glycine, The massive CySyn polypeptide represents the upper limit of molecular size of the NRPS enzymes. A molecular mass of 1.69 MDa (15 281 amino acids), was delineated from the sequence of the CySyn gene, simA, which constitutes an intronless genomic open-reading frame (ORF) of 45.8 kb (Weber et al. 1994; Velkov and Lawen 2003). The role of this gene in CyA biosynthesis was proved by Weber and Leitner (1994). They demonstrated that the knock-out of the simA gene in Tolypocladium inflatum resulted in its inability to produce cyclosporins. Several members of Cy family (like CyA) contain nonproteinogenic amino acids (D-alanine, Abu and unusual Bmt or a similar C9-amino acid; Fig. 8.2), which have to be synthesized by a pathway independent of the primary metabolism. Therefore, besides CySyn, the presence of some other enzymes is crucial for CyA biosynthesis as MeBmt L1 MeVal L11 MeLeu L10 CH3 Abu L2 CH3 CH3 H3C H3C H3C CH3 O CH3 HO N CH3 N O H3C CH3 N CH3 O MeLeu L9 O H N H N CH3 O OH C 3 H3 C CH3 O O D-Ala L8 CH3 H N H N H3C N O N Sar L3 N H N CH3 L-Ala L7 O O CH3 H3C MeLeu L6 H3C CH3 CH3 Val L5 MeLeu L4 Fig. 8.2. The chemical structure of cyclosporin A including the numbering system. It is composed of 11 amino acid unit, with seven of the amide nitrogen methylated. The three non-proteogenic amino acids are: D-alanine, Abu (L-2 aminobutyric acid) and Bmt (4R)-4-[(E)-2-butyl]-4-methyl-L-threonine (modified from Velkov et al. 2006) 170 Hesham El Enshasy Transmission electron micrographs of negatively stained CySyn macromolecules showed large globular complexes of 25-30 nm in diameter, built up by smaller interconnected units associated with smaller particles of 7 nm length. Complexes of CySyn and D-alanine racemase are linked and localized at the fungal vacular membrane, where Cy synthesis is carried out (Hoppert et al. 2001). CySyn and D-alanine racemase seem to be located in close vicinity to each other, since D-alanine is the leading amino acid of the polypeptide chain synthesized by CySyn. Cyclosporin is subsequently accumulated inside the vacuoles and released slowly through vacuolar and cytoplasmatic membranes or rapidly upon cell lysis. CySyn was prepared in purified form at pilot scale and used as a model to produce large amounts of CyA in vitro. The process included ammonium sulfate precipitation, gel filtration, hydrophobic interaction chromatography and anion exchange chromatography, and it yielded an electrophoretically homogenous cyclosprin synthetase preparation (Velkov et al. 2006). The obtained enzyme exhibited an optimal temperature range between 24 and 29 ºC and a pH optimum around 7.6. 3. Production The production of cyclosporins at the laboratory scale can be carried out using different aerobic filamentous fungi such as Tolypocladium inflatum, Fusarium solani (Sawai et al. 1981), Neocosmospora vasinfecta (Nakajima et al. 1989), Acremonium luzulae (Moussaı̈f et al. 1997) and T. cylindrosporum (Sekar et al. 1997). The industrial production of cyclosporins is mainly performed using highly productive strains of T. inflatum. This organism was originally mis-classified as Trichoderma polysporum Gams, however, later it turned out that it belonged to a new genus of ascomycetous molds, Tolypocladium and coined the name T. inflatum (Gams 1971). In 1983, another research group found that T. inflatum was identical to Pachybasium niveum, and since the latter older name has priority under the rules of the International Code of Botanical Nomenclature, the strain was renamed as T. niveum (Bissett 1983). This fungus was again re-classified as Beauveria nivea (Von Arx 1986). Based on the research of Kathie Hodge, this strain was found to be the asexual state of Cordyceps subsessilis (Hodge et al. 1996). Due to the economic importance of this fungus, the classification as T. inflatum was neverteless conserved for cyclosporin producers to avoid any confusion with other strains (Dreyfuss and Gams 1994). In spite of some efforts to produce CyA with immobilized cells or by solid-state fermentation (SSF), the industrial production of this immunosuppressive agent is mostly carried out using free cells in submerged cultures in stirred-tank bioreactors. The particular role of the type of strain on the production of CyA along with some characteristic morphological features was reported by several authors. In case of T. inflaturm, large intra-population variations in colony color and shape were observed on solid media. Thus, colony color can range from white to brownish (including yellow, orange and red colonies) (Aarnio and Agathos 1990). The production of a pink pigment was found to be associated with cyclosporin production in certain T. inflatum strains (Chun and Agathos 1989). Besides the selection of highly productive colonies of wild-type strains, attempts were undertaken to increase the strain productivity by mutation using chemical mutagens such as methyl sulphate, epichlorohydrin or nitrosoguanidine (Agathos et al. 1986). A recent study of Mi-Jin and coworkers (2009) has demonstrated the possibility of the improvement of T. niveum productivity by using random mutagenesis combined with protoplast transformation. The mutant strain, generated using a random UV method, produced more than ninefold higher amounts of CyA than the wild-type strain. Additionally, a bacterial gene of a Vitreoscilla spp. (hemoglobin gene, VHb) was transferred to the UV-irradiated mutant to increase oxygen uptake in liquid culture and led to an additional increase in CyA production of more than 30%. Besides the type-strain used, the production levels of CyA are dependent on several regulating factors such as inoculum type and size, medium composition and additives as well as process parameters such as temperature, pH and partial oxygen pressure. A high density of the sporeinoculum was found to be necessary for the development of small pellets, which is the preferred morphology for cyclosporin production (Dayfuss et al. 1976; Isaac et al. 1990). However, inoculum size is only one of more than 20 other factors controlling the fungal pellet formation (El Enshasy 2007). The influence of the type and concentration of carbon and nitrogen sources on CyA production has been examined in wild-type and mutant strains of T. inflatum. Among different carbon sources tested, 3% sorbose gave the highest CyA titre (Agathos et al. 1986). A feeding strategy using the sequential addition of two carbon sources (sorbose and maltose) was also reported to be successful in attaining a higher volumetric production (Agathos et al. 1986). Another study showed that an optimal medium for CyA production can be developed by factorial experimental design and consisted of the three carbon sources glucose, sucrose and starch in different ratios (Abdel Fattah et al. 2007). Biosynthesis of CyA was found to be heavily influenced by the external addition of amino acid Immunomodulators constituents of the molecule. Addition of L-valine increased the specific production of CyA by 60% in semi-synthetic media and even by 400% in synthetic media. Experiments using repeated addition of L-valine indicated that the amino acid has to be present in the exponential growth phase of the fungus for optimal CyA production (Lee and Agathos 1989). Based on this finding, a mathematical model for the production of CyA in the presence of supplemented L-valine was developed, which also considered kinetic information and mechanistic data on CyA biosynthesis (Agathos and Lee 1993). Concomitant addition of L-leucine and L-valine to a synthetic medium was found to stimulate CyA production as well (Balakrishnan and Pandey 1996). When fungal cells enter the stationary phase and CyA accumulates in the medium, they partially undergo lysis and CyA degradation sets in, especially under carbon source limitation. The intensity of cell lysis and CyA degradation in the bioreactor was higher than in agitated flasks, especially under an uncontrolled pH regime (El Enshasy et al. 2008). Several attempts have been made to use immobilized cells for CyA production. On example is the successful production of CyA in high amounts using carrageenan-entrapped cells of T. inflatum in an airlift bioreactor (Foster et al. 1983). CyA was also produced in relevant amounts by a Tolypocladium sp. immobilized in calcium alginate beads in a packed-bed reactor (Sekar and Balaraman 1998a). Continuous production of CyA was realized using immobilized spores of T. inflatum on celite beads (Chun and Agathos 1989). The CyA productivity by cells immobilized on celite beads (100–500 mm) was reported to be 4–6 mg l–1 h–1. This value is about six- to tenfold higher than those of batch fermentations in suspension cultures (Lee et al. 1997). Furthermore, attempts were made to produce CyA by solid-state fermentation (SSF) to reduce the production costs. So in a study, wheat-bran was used as a solid support and yielded up to 1400 mg CyA kg–1 substrate (Sekar et al. 1997). After optimizing different cultivation parameters, such as the type and design of tray, thickness of the solid substrate bed, type and size of inoculum as well as relative humidity, the CyA production increased to a value of 1920 mg kg–1 (Sekar and Balaraman 1998b). However, SSF up-scaling raises severe engineering problems due to difficulties of adjusting temperature, pH, oxygen and moisture content as well as of managing gradient formation inside the cultivation system. Recently, a novel process for CyA production by F. solani using a large-scale SSF bioreactor of an area of 226 m2 has been developed (Khedkar et al. 2007). Besides the continuous optimization of the up-stream part of the production process, improvement of CyA extraction methods has also contributed to increase the overall yield of the process. CyA is a hydrophobic molecule with high solubility in 171 low-molecular-weight alcohols, and extraction could be optimized using different alcohols as solvent system and varying temperatures during the extraction process (Ly and Margaritis 2007; Ly et al. 2007). 4. New Generations A recent study has demonstrated that CyA has a number of side-effects causing among others hypertension, dyslipidemia, hirsuitism and chronic renal insufficiency that leads in 10% of cardiac transplant recipients to an end-stage renal disease (Patel and Kobashigawa 2008). Numerous analogues and derivatives of CyA have been tested in order to improve the drug’s therapeutic properties. For example, CyG, a cyclosprin A analogue with a L-novaline substituent at position 2, displays equal immunosuppressive effects as CyA but with less nephrotoxicity (Hiestand et al. 1985). Another derivative SZZ IMM-125, which is a hydroxyethyl derivative of D-serine-8-cyclosporine, was found to be slightly more potent but far less nephrotoxic than CyA in both in vitro and in vivo models (Hiestand et al. 1992; Ferraresso and Kahan 1993). ISATX247 is a potent derivative with higher activity and lower nephrotoxicity compared to CyA (Gregory et al. 2004). Several other cyclosprin analogues with high immunosuppressive activity were obtained through the chemical modification of the side chains at the first amino acid and optionally at third amino acid (Molino and Yang 2006). B. Mycophenolic Acid Mycophenolic acid (MPA), [6-(4-hydroxy-6methoxy-7-methyl-3-oxophthalanyl)-4-methyl-4hexenic acid; CAS 24280-93-1; Figs. 8.1–8.2], is one of the oldest known secondary metabolites. The compound was first detected in 1896 by Gozio in the fermentation broth of Penicillium glaucum and recognized as a lipid-soluble weak organic acid. This compound was also isolated from the culture filtrate of Penicillium stoloniferum Thom by Alsberg and Black (1913), who named it MPA (Alsberg and Black 1913; Jekkel et al. 2001). Since then, numerous reports have been published dealing with the production of MPA by different microorganisms. The complete chemical structure of MPA was first reported by Birkinshaw et al (1952). OH CH3 OH CH3 HO HO2C O O methylphthalide + 3 CH2COSCoA CH3COSCoA HO2C O Fig. 8.3. Different steps of MPA biosynthesis O CH3 O HO 2 OH CH3 OH 5-methyl-orsellinic acid SCOA HO CH2 Demythylmycophenolic acid (DMPA) O O CH3 OH CH3 CO2H HO 5,7-dihydroxy-4-methylphthalide O O HO2C Mycophenolic acid O O CH3 H3CO 6-farnesyl- 5,7-dihydroxy-4- O CH3 HO 3 OH CH3 CH3 The MPA 2-morpholinoethyl ester (also named mycophenolate mofetil, MMF) of the chemical structure 2-morpholinoethyl-(E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl3-oxo-5-isobenzofuranyl)-4-methyl-4-hexeneoate (CAS 128794-94-5; Figs. 8.1–8.3), is one of the most important MFA derivatives and approved by the FDA in 1995 as immunosuppressive agent for the prevention of acute renal allo-graft rejection and in 1998 for heart transplantation. A further improved generation of MPA drugs, based on sodium mycophenolate in controlled release formulations with good gastrointestinal absorption and bioavailability, was approved in 2005 (Xy and Yang 2007). In addition to the well established market for organs transplantations, MPA and its derivatives have been recognized by many physicians as an effective option for the treatment of immune-mediated diseases (Bentley 2000; Mydlarski 2005). Another pro-drug of MPA was developed in Japan during the early 1980s, initially as antitumor agent against various experimental cancers (Mitsui et al. 1981; Matsuzawa and Nakase 1984). In this derivative, the hydroxyl group of MPA was derivatized and the carboxylic functionality was replaced by an ethyl ester to produce ethyl-[N-(p-carboxyphenyl)-carbamoyl]-mycophenolate, abbreviated as CAM. The chemical structure of CAM was fully characterized and its crystal structure solved by Nawata and coworkers (Nawata et al. 1988; 1989). An early study using CAM as immunosuppressive drug demonstrated that it can suppress acute allergic cephalomelitis in Lewis rats (Mizobuchi et al. 1997). Based on this study, it was suggested that CAM might be also a useful adjunct for the long-term immunosuppressive therapy of inflammatory diseases of the central nervous system. At the same time, a study of Sawada and his group demonstrated the usefulness of CAM in bowel transplantation (Sawada et al. 1996). Using a rat model, the immunosuppressive activity of CAM was explained through the inhibition of the interphotoreceptor retinoid-binding protein (IRBP) mediated autoimmune CH3 At the early stage of its biomedical applications, MPA was used as a broad-spectrum antibiotic due to its antibacterial, antifungal, antiviral and antiprotozoal activities (Abraham 1945; Ando et al. 1968; Cline et al. 1969; Noto et al. 1969). Moreover, it was found to exhibit also some antitumor and antipsoriasis as well as anti-inflammatory activities (Carter et al. 1969; Spatz et al. 1978; Epinette et al. 1987). MPA was produced during that time under the trademark Bialin (Vinkurova et al. 2005). However, it has not widely used in practice as antibiotic because most microbes were found to readily become resistant to this compound. In spite of its low acute toxicity to mammals, MPA was also described as a mycotoxin by some authors (Sanchis et al. 1988; Puel et al. 2005). New interest in MPA and its derivatives has remarkably grown after the discovery of their immunosuppressive properties. O Hesham El Enshasy C 172 Immunomodulators uveoretinitis by a decrease in cytokine production (Sakai et al. 1999). Compared to other immunosuppressive drugs, CAM has only minor adverse side-effects due to its relatively specific action on lymphocytes. Furthermore, this new derivative was more effective than MMF in prolongation of heart-graft survival in rats at each dose applied (Takazawa et al. 1995). Nowadays, MPA derivatives used as immunosuppressant drug in organ tansplantation are marketed under different trade names, such as CellCept (mycophenolate mofetil, Roche) and Myfortic (mycophenolate sodium; Novartis). In case of oral applications of MMF, the pro-drug mycophenolate mofetil is rapidly hydrolyzed to MPA after administration and suppresses the immune system via a non-competitive reversible inhibition of inosine-5’-monophosphate dehydrogenase (IMPDH; EC 1.1.1.205). This enzyme catalyzes the NAD-dependent oxidation of inosine-5’-monophsphate (IMP) to xanthosine-5’monophosphate (XMP), which is the committed step in the de novo biosynthesis of guanosine monophosphate (GMP). This reaction is particularly important to generate the guanosine nucleotide levels needed to initiate a proliferation response of B- and T-lymphocytes to mitogens and antigens (Sintchak et al. 1996). Thus, MPA acts as a potent anti-proliferative agent (Hood and Zarembski 1997) affecting cytokine-dependent signals and causing in vivo the inhibition of lymphocyte reactions (Allison and Eugui 2000). In consequence, MPA and its derivatives are being widely used in the transplantation of different organs in humans at different ages (Budde et al. 2006; Tönshoff 2006; Aw et al. 2008). Besides the wide application of MPA in organ transplantation, it is also used in the treatment of immune related diseases such as rheumatoid arthritis, lupus inflammatory bowel disease and other kidney or skin disorders (Liu and Mackool 2003; Appel et al. 2005; Hartmann and Enk 2005; Iaccarino et al. 2007). Moreover, MPA has recently been used in the treatment of rare diseases like interstitial nephritis (Preddie et al. 2006) and focal segmental glomerulosclerosis (Cattran et al. 2004). In general, MMF has several advantages over cyclosporins as maintenance therapy of organgraft recipients (Eugui and Allison 1993). Above all, MMF is well tolerated by the human body and has a lower toxicity and hence fewer side-effects than CyA. 173 1. Chemistry Already before the discovery of its immunosuppressive activity, MPA was used as drug due to its wide biological activity against bacteria, parasites and viruses. Thus for many years modifications of the MPA structure were the subject of intensive research in order to increase its biological activity, bioavailability and the range of applications (Lee et al. 1990; Nelson et al. 1990; Rohloff et al. 1995). Most studies focused on increasing the antitumor activity of MPA through the production of monocyclic analogues and carboxamide derivatives without any change in the aromatic ring and the surrounding side chains, since the free phenolic structure is an absolute prerequisite for MPA activity (McCorkindale and Baxter 1981; Anderson et al. 1996; Menza-Aviñ et al. 2005). The methoxy and methyl groups of the aromatic ring represent two other key structural elements influencing the activity of MPA (El-Araby et al. 2004). The study of Nelson et al. (1996) demonstrated that the aromatic methyl group of MPA is essential for its biological activity and the replacement of the methoxyl group by other ethers resulted in compounds with two- to fourfold higher potency in vitro and in vivo. Further improvement of MPA activity was achieved by the development of new MPA analogues, which have overcome the drawback of glucuronidation of the phenolic hydroxyl group at C7 (Chen et al. (2007). In these derivatives, a truncated MPA is connected to an adenosine moiety via a linker (e.g. methylene bis-phosphonate) leading to mycophenolic adenine dinucleotide derivatives. The new molecules show a better biological activity and chemical stability. Moreover, a new series of IMPDH inhibitors based on the replacement of the benzofuranone moiety in MPA by a methoxy-(5-oxazolyl)-phenyl (MOP) moiety has recently been developed (Chen et al. 2008). Besides the different methods of chemical modification, several attempts were made to transform MPA using different microorganisms (Jekkel et al. 2001). In the course of these bioconversions, mycophenolic acid was found to undergo one or more of the following transformations: hydroxylation at the side chain or the lactone ring, amide or alcohol formation at the carboxylic acid group, oxidative cyclizations of the side chain or glycosylation (Jekkel et al. 2002). 174 Hesham El Enshasy 2. Biosynthesis The molecule of MPA consists of an acetatederived aromatic nucleus, a terpenoid side chain, and two methyl groups (the 5-methoxyl and the 4methyl group). Different schemes of MPA biosynthesis have been proposed by different authors (Bedford et al. 1973; Muth and Nash III 1975, Nulton and Campbell 1978). In 2000, Bentley summarized and updated the synthesis pathway in his excellent review (Fig. 8.3). Accordingly, MPA biosynthesis involves two major pathways of secondary metabolite formation: the polyketide and the isoprenoid pathway as well as methylation reactions on oxygen and carbon atoms. In this process, a typical acetate-polymalonate condensation with a methylation prior to condensation leads to the aromatic structure, 5-methyl-orsellinic acid. It proceeds thereafter through lactone formation followed by addition of the C15-farnesyl diphosphate unit and formation of the intermediate 6-farnesyl-5,7-dihydroxy-4-methylphthalide (Fig. 8.3). This compound undergoes several oxidative degradation steps to remove eight carbon atoms from the side chain by two possible mechanisms: one involves two oxidative cleavages at the two side chain double bonds removing levulinic acid and acetone from the aromatic ring. The other possible mechanism is a direct oxidation at the central double bond of the farnesyl side chain. The two pathways are regarded as being of equal importance (Bentley 2000). Early studies had already shown that the basic carbocyclic skeleton of the molecule was acetatederived and that methionine provided the O- and C-linked methyl groups attached to the aromatic ring (Birch et al. 1958; Jaureguiberry et al. 1964). The last step of MPA biosynthesis was found to be the transfer of a methyl group from S-adenosyl-L-methionine (SAM) to demethylmycophenolic acid (DMPA). This step is catalyzed by a specific SAM:DMP O-methyltransferase (Muth and Nash 1975). 3. Production MPA was originally isolated from culture filtrates of Penicillium glaucum and P. stoloniferum as a weak acid with antifungal activity (Alsberg and Black 1913); later MPA production was reported for 12 strains of the species P. brevicompactum (Clutterbuck et al. 1932). Since that time, many reports have been published dealing with the production of MPA using different species of the genus Penicillium (Vinokurova et al. 2005), such as P. brevicompactum (Doerfler et al. 1979; Ozaki et al. 1987a; Alani et al. 2009), P. brunneostoloniferum (Nakajima et al. 1979); P. roqueforti (Lafont et al. 1979, Engel et al. 1982; Schneweis et al. 2000) and other molds like Neocosmospora spp. and Byssochlamys nivea (Puel et al. 2005). Unfortunately, most of the studies on MFA production were carried out on a small scale and data on the detailed effects of media components and cultivation conditions are scarely found in the literature. Among different MPA producers, the highest productivity was obtained for strains of P. brevicompactum and P. stoloniferum which both are suitable for industrial fermentation (Queener and Nash 1978; Kida et al. 1984; Sircar et al. 2005). Several attempts have been made to improve MPA production by using antibiotic-resistant mutants with a high internal ergosterol level (Queener et al. 1982). Also, rational breeding procedures based on the biosynthetic pathway were used to select strains with improved MPA productivity. Among the different antibiotic-resistant mutants developed, a clofibrate and dodecyltrimehylammonium chloride double resistant mutant produced about 4.7 g l–1 MPA (about three times more than the parent strain, P. brevicompactum ATCC 16024). A glutamate auxotroph of this antibioticresistant mutant was even able to produce up to 5.8 g l–1 MPA. This strain was found to grow on L-aspartate instead of L-glutamate and exhibited only one-third of the pyruvate carboxylase activity of the parent strain (Ozaki et al. 1987a). Cultivation in submerged culture showed that the production of MPA starts concomitantly with the hyphal aggregation phase just before pellet formation (Doerfler et al. 1978). Unlike most secondary metabolites, MPA is produced growth-associated in the exponential phase (in both batch and continuous cultures) and independent of the medium composition (Nulton and Campbell 1977; Doerfler et al. 1979). The production process is carried out either in submerged cultures or by solid-state fermentation (SSF). For many years, the optimal medium for MPA production has been a semi-synthetic mixture composed of glucose (C-source), ammonium salts or casein (N-source), potassium dihydrogen phosphate (P-source), magnesium sulphate and trace elements. Some authors supplement other components like the amino acid glycine (Xu and Yang 2007) to further increase MPA production. More recently, it has been reported that, among different nitrogen sources, urea in concentrations up to 5 g l–1 was the Nsource of choice to support MPA production (Roh 2008). However all in all, only little efforts haven been done to optimize media composition and cultivation conditions compared to the production of other important fungal metabolites. Immunomodulators Like other production processes involving fungal cells, growth morphology is a critical factor determining the growth rate and production yield. Fungi can grow either in form of pellets or mycelia and thus, controlling the growth morphology to a desired shape is important to improve the cell productivity. Altogether, more than 30 factors have been reported in the literature to influence the growth morphology; these include straindependent factors (type of strain, inoculums size, physiology, etc.), cultivation conditions (pH, temperature, osmotic stress, etc.) and medium composition (C-source, N-source, C/N ratio, surfactants, presence of insoluble particles) and many other factors (El Enshasy 2007). Using spores as inoculum for MPA production in submerged culture, it was shown that the increase of spore density from 104 to 107 spores ml–1 resulted in significant reduction in pellet size with a concomitant increase in MPA production from 0.2 up to 4.8 g l–1. Further, it was found that MPA can be continously produced, independent of spore concentration, in the presence of 1% celite in the culture medium. In the case of celite addition, growth occurred in form of pellets (500 mm in diameter) regardless of the inoculated spore concentration (Ozaki et al. 1987b). New cultivation approaches try to overcome the problem of changed cell morphology by applying rotating fibrous-bed bioreactors (RFB). Accordingly immobilized cells in RFBs produced MPA up to a concentration of 5.7 g l–1 within 14 days using the standard wild-type strain P. brevicompactum ATCC 16024. Other advantages of RFB fermentation include the ease of product separation and purification from the fermentation broth as well as the possible repeated use of cells for long-term operation (Xu and Yang 2007). Several attempts were made to use solid-state fermentation (SSF) as an alternative cultivation method. In general, filamentous fungi are well suited for SSF and a number of valuable metabolites can be produced under these conditions, since they perfectly reflect the natural habitats of the fungi (Krishna 2005). 175 (2009) has demonstrated that MPA production by SSF in a packed-bed bioreactor can lead to yields up to 6900 mg MPA kg–1 pearl barley within just 168 h. Due to its low molecular weight and the relatively simple chemical structure, MPA and its derivatives can nowadays also prepared chemically. Different procedures for MPA total synthesis have been published using different starting materials. Patterson (1993) synthesized MPA using silyloxy-1,3cyclohexadiene and allylic alcohol via an ortho-ester Claisen rearrangement. Another interesting method for a convergent synthesis of MPA via a palladium-tin coupling reaction between the alkyl side chain and the phthalide ring was described by Plé et al. (1997). A further method involves 2-geranyl-1,3-acetonedicarboxylate and 4-pivaloyoxy-2-butynal and a specific cyclization as key step (Covarrubias-Zúñiga and Gonzlez-Lucas 1998). The production of MPA using this new synthetic strategy is based on a ring annulation sequence involving a Michael addition reaction and an intra-molecular Dieckmann condensation in situ (Covarrubias-Zúñiga et al. 2003). C. Mizoribine The immunosuppressive antibiotic mizoribine or bredinin (5-hydroxy-1-b-D-ribofuranosyl1H-imidazole-4-carboxamide; CAS 50924-49-7; Fig. 8.4) was first isolated from the culture medium of Eupenicillium brefeldianum isolated from soil samples on Hachijo island (Japan; Mizuno et al. 1974). Mizoribine (MZA) is an imidazole nucleoside and the metabolite MZ-5-P exerts its activity through selective inhibition of inosine monophosphate synthetase and guanosine monophosphate synthetase, resulting in the complete inhibition of guanine nucleotide biosynthesis (Shumpei 2002). Based on this immunosuppressive mechanism, mizoribine is superior to many O An early study by Bartman et al. (1981) demonstrated that, when cells grew as surface culture, MPA production was associated with the aerial mycelium and its production ceased completely when the formation of aerial hyphae was blocked. However, the yield of MPA in this study was relatively low (only 0.3 mg g–1 wet weight). SSF production of MPA was optimized using a response surface methodology and the maximal yield achieved was 3300 mg kg–1 wheat bran (Sadhukhan et al. 1999). Further improvement was achieved by using a fed-batch strategy (Tiwari et al. 2003). Furthermore, a recent study of Alani et al. NH2 N OH N OH O OH Fig. 8.4. Mizoribine OH 176 Hesham El Enshasy other clinically used drugs, since it may not cause damage to normal cells and nucleic acids. In contrast to other immunosuppressive agents widely used at the time of its discovery (e.g. azathioprine), mizoribine was shown in animal experiments to lack oncogenicity and exhibited a clinically low incidence of side-effects such as hepatotoxicity and myelosuppression. These facts together supported its use in clinical application in long-term immunosuppression therapies. In 1984, MZR was first approved for the treatment of graft rejection after kidney transplantation (Takei 2002). Later, it was also approved for the treatment of other diseases including lupus nephritis, rheumatoid arthritis and primary nephritic syndrome. It is currently marketed in China, Korea and Japan under the trade name Bredinin (Tanaka et al. 2006). The drug is mainly produced by the fungi E. brefeldianum and E. javanicum in submerged culture under aerobic conditions, however, little information is available on the biosynthesis pathway and the production process (Mizuno et al. 1975; Benedetti et al. 2002). D. Other Immunosuppressants In addition to the three clinically approved fungal immunosuppressants, Cyclosporins, MPA and mizoribine, many other fungal metabolites possess also potent immunosuppressive activities (Fig. 8.5). Some of them were found to be not suitable for clinical applications due to their side-effects, whereas others are still subject of intensive studies and currently undergoing different levels of clinical trials till final approval by FDA. 1. Ovalicins and Fumagillins The fungal metabolites ovalicin, fumagillin and their related derivatives belong to the most potent anti-angiogenic compounds. They bind convalently to the active site of the enzyme methionineaminopeptidase type 2 (MetAP2) and irreversibly block its proteolytic activity (Liu et al. 1998; Turk et al. 1998). Ovalicin or Graphinone (CAS 19683-98-8) is a sesquiterpene that was first isolated from culture filtrates of Pseudeurotium ovalis in 1962 and found to have antimicrobial and cytotoxic activities. It was chemically characterized by Sigg and Weber (1968). The chemical structure of this compound is related to the antibiotic fumagillin. The immunosuppressive properties of ovalicin were evaluated using the mouse hemagglutinin test, which reflects the degree of antibody production. It was found be not toxic to the cells of bone marrow, which distinguished it from existing immunosuppressants at that time. Unfortunately, when tested in humans it later turned out to have other toxic side-effects. A Metarhizium sp. isolated from soil in Japan was found to produce a 12-hydroxyovalicin (Kuboki et al. 1999), which was named Mer-f3. This compound was examined for its influence on mixed lymphocyte cultures, and showed a similar inhibitory activity as ovalicin. Mer-f3 had an immunosuppressant activity in the murine mixed lymphocyte test with IC50 ¼ 1 nM which is even better than that of CyA (110 nM). Moreover, Mer-f3 had no inhibitory activity on leukemia L-1210 which indicates a low mammalian toxicity. Another Metarhizium sp. isolated from soil was reported to produce a novel immunosuppressive substance, which was named metacytofilin (Iijima et al. 1992); this compound had not any antimicrobial activity, while showing strong immunosuppressive effects. Chlovalicin, a chlorinated compound derived from the epoxide ring attached to ovalicin, was discovered in the fermentation broth of the soil fungus Sporothrix sp. It inhibited the IL-6 dependent growth of MH60 cells and appeared to be a new IL-6 inhibitor (Hayashi et al. 1996). Two ovalicin related compounds, FR 65814 (CAS 10347060-6) and Fumagillol (CAS 108102-51-8), were isolated from culture filtrates of the soil fungus Penicillium jensenii. They both showed significant immunosuppressive activity at low concentrations (Hatanaka et al. 1988). A chiral and stereoselective total synthesis of FR 65814 using glucose as starting material was described by Amano et al. (1998, 1999), and fumagillol can be also synthesized using other starting materials (Kim et al. 1997, 2005a; Boiteau et al. 2001). TNP-470, a semisynthetic derivative of fumagillin, reduced the proliferation of endothelial cells with an IC50 value of 2.510–11 M. Therefore, it entered clinical trials as immunosuppressive and anti-tumor agent. The main drawbacks of its therapeutic properties were the short physiological half-life span and the severe side-effects such as ataxia, vertigo and agitation (Figg et al. 1997). The relation between the chemical structure and bioactivity of fumagillin and its derivatives was studied by several authors. It was shown that the spiro-epoxide is essential for the activity of fumagillin and its conversion into a methylene group results in a considerable reduction of eficacy (Logothetis et al. 2001). On the other hand, the epoxide of the side chain has no major effect on the biological activity. New synthetic analogues of ovalicin and fumagillin lacking reactive epoxy functionalities, which are thought to be responsible for the severe toxic side-effects, were synthesized by Mazitschek et al. (2005). 2. Gliotoxin Gliotoxin (CAS 67-99-2) is a sulfur-containing antibiotic that belongs to the epipolythiodioxopiperazine group of secondary metabolites and Immunomodulators R CH3 O CH3 O CH3 CH3 O CH3 Ovalicin Ovalicin :R=H : R = OH CH3 OH Fumagillin CH3 CH3 O N OCH3 O S S CH3 N OH OH OH Cl O O 4 Fumagillin TNP470 CH3 CH3 O NH O CH3 HO OCH3 O O Fumagillin CH3 O O H OCH3 OH O CH3 O OH OCH3 CH3 CH3 O OH O 177 Gliotoxin CH3 OH OH OH OH CH3 CH3 CH3 H3C H3C H3C H OCH3 OCH3 OCH3 Flavidulols Flavidulols Flavidulos OCH3 OH OH HO CH3 NH2 O OH O Myriocin CH3 OH NH2 OH Myriocin Fig. 8.5. Molecular structure of different types of fungal immunosuppressive agents exhibits antifungal, antiviral as well as strong immunosuppressive activities. Gliotoxin was originally isolated from Gliocladium fimbriatum and named accordingly. It was reported thereafter that this compound is commonly produced by several genera of molds such as Aspergillus, Trichoderma and Penicillium. Gliotoxin was also claimed to be produced in yeasts of the genus Candida, however, a recent study being based on a screening of 100 clinical isolates of Candida doubted the occurrence of this compound in yeasts (Kupfahl et al. 2007). The immunosuppressive effects of gliotoxin have been explained by different mechanisms. It suppresses cell activity and induces apoptosis in a variety of cell types including neutrophils, esosinophils and granulocytes (Ward et al. 1999). It inhibits the chymotrypsin-like activity of the 20S proteasome in a non-competitive manner (Kroll et al. 1999) and likewise the activation of NF-kB in T- and B-cells when applied at nanomolar concentrations (Pahl et al. 1996). The immunosuppressive activity of gliotoxin was also attributed to the inhibition of perforin-dependent and Fas-ligand-dependent cytotoxic T-lymphocyte-(CTL)-mediated cytotoxicity (Yamada et al. 2000). Another study has furthermore demonstrated that gliotoxin suppresses the mast cells, which play a key 178 Hesham El Enshasy O O H N Pro H3C CH3 H Ala-Alb-Alb-X-Ala-Alb-Y NH H MDA H CH3 H OH H O H3C N CH3 O H H3C OH HO H H3C HMDA Trichopolyn Acyl group I II III IV V X group MDA MDA MDA MDA HMDA Ileu Val Ileu Val leu OH OH CH3 H H3C H H3C O OCH3 Kobifuranone A H3C Kobiin OH R 1 OH CH3 O O Kobifuranone B Kobifuranone C H2N R=H R=OH Terprenin 3-Methoxyterprenin 4'-Deoxyterprenin 1 2 : R = OH , R = OH 1 2 : R = OCH3 , R = OH 1 2 : R = OH , R = H OH HO CH3O HO Mycestericin A O H3CO R CH3 R2 O CH2 CH3 OH CH3 CH3 HO Y group α-aminoisobutyric acid α-aminoisobutyric acid Ala Ala α-aminoisobutyric acid O R HO O P OH OH R= A CH3 HO H B N C Mycestericin B R= CH3 NHCH3 HO H Mycestericin C R= CH3 FR901483 O Fig. 8.5. (continued) role in host defense and are important in both innate and adaptive immunity (Niido et al. 2006). As this compound is produced by many potential human pathogens in vivo during the course of infection, gliotoxin may also contribute to the etiology of fungal diseases (Waring and Beaver 1996). 3. Trichopolyns Trichopolyns (TPs) are peptabiotic compounds produced by Trichoderma polysporum. TPs I and II were first isolated as new antifungal and antibacterial antibiotics in 1978 by Fuji and coworkers. The chemical structures of TP I (CAS 66554-87-8) and TP II (CAS 6655-88-9) were identified as peptide antibiotics three years after their discovery (Fujita et al. 1981). Nowadays, the structures of five different trichopolyns (I–V) are known. Trychopolyns I and II are ten-residue peptides characterized by the presence of a 2methyldecanoyl group at the N-terminus, and the C-terminal residue is protected by trichodiaminol. The other three analogues, TPs III-V differ from TPs I and II in that way that Aib (a-aminoisobutyric acid) is replaced by L-alanine. In contrast, TP V has the same amino acid sequence as Immunomodulators TP I, but the N-terminal acyl group is substituted by 3-hydroxy-2-methyldecanoic acid (instead of 2-methyldecanoic acid). These peptabiotics have been shown to suppress the proliferation of lymphocytes in mouse allogeneic mixed lymphocyte reactions (Lida et al. 1999). The TP I activity was even stronger than that of CyA. 4. Myriocin Myriocin (antibiotic ISP-1 or thermozymocidin; CAS 35891-70-4) was first isolated from the thermopholic fungus Myriococcum albomyces by Kluepfel and his group in 1972 and patented in the United States in 1975 (Kluepfel et al. 1975). The compound was recognized as an active antibiotic against yeasts and dermatophytes when applied in vitro. However, the compound appeared to be too toxic for therapeutic purposes in humans. More than 20 years later, the same compound and its derivatives (mycestericins) were isolated from Isaria sinclairii, which is the imperfect stage of Cordyceps sinclairii, and showed strong immunosuppressive activities. Myriocin was 10- to 100-fold more effective than cyclosporin A both in in vivo and in vitro tests (Fujita et al. 1994; Sasaki et al. 1994). Isaria sp. belong to the entomopathogenic fungi colloquially called “vegetable wasps and plant worms”, which have been used in oriental medicine for more than 1000 years (Im 2003). Myriocin was found to suppress both the production of antibodies against red blood cells of sheep and the induction of cytotoxic T-lymphocytes more strongly than cyclosporine A. It is also a potent inhibitor of serine palmitoyltransferase (SPT), the enzyme that catalyzes the first step in sphingosine biosynthesis (Miyake et al. 1995). Thus, it is used in biochemical research as a tool for depleting cells of sphingolipids. Myriocin can also be produced by chemical methods (Banfi et al. 1982; Oishi et al. 2002; Jones and Marsden 2008). Fingolimod or FTY720 (CAS 162359-55-9) is a novel immunosuppressant obtained by chemical modification of myriocin (Adachi et al. 1995). A number of alternative ways for its preparation using shorter pathways for synthesis with higher overall yields have been published over recent years (Seidel et al. 2004; Adachi and Chiba 2007). Fingolimod was actually designed to eliminate the GI toxicity of the original compound myriocin. The exact mechanism of its immunosuppressive activity, however, remains still unclear. Some researchers have hypothesized that FTY720 may induce the apoptosis of lymphocytes (Suzuki 1996; Fujino et al. 2002). Others have proposed that the number 179 of lymphocytes decreases as a result of their movement towards secondary lymphoid organs such as lymph nodes and peyer’s patches (Sugito et al. 2005). It is certain that this novel immunosuppressant prolongs the survival of allo-graft transplants and is effective in the treatment of some immunological diseases. At the moment, FTY720 is being further developed by Novartis in phase II clinical trials. Not least, this compound may have a great clinical potential because of eficacy as oral drug for the treatment of multiple sclerosis (Gullo et al. 2006; Klatt et al. 2007). 5. Flavidulols The immunosuppressive geranylphenols, flavidulols A (CAS 117568-32-8), B (CAS 117568-33-9) and C (CAS 117568-34-0), were originally isolated from fruiting body extracts of the mushroom Lactarius flavidulus in the course of a screening for new inhibitors of the proliferation of mouse lymphocytes (Takahashi et al. 1988). The chemical structure of these compounds was determined by NMR analysis (Takahashi et al. 1993); this paper also reported on the isolation of a new flavidulol D (CAS 156980-40-4). The suppressive effects of flavidulols A, B and C on the proliferartion of mouse lymphocytes were stimulated in the presence of mitogens such as concavalin A (CoA) and lipopolysaccharides (LPS). Their IC50 values for the inhibition of mitogen-induced concavalin A proliferation of mouse lymphocytes were between 9 and 36 mg ml–1 and against lipopolysaccharideinduced proliferation between 7 and 28 mg ml–1 (Fujimoto et al. 1993). 6. Kobiins The sesterterpenetriol immunosuppressant kobiin and another three related 2-furanones named kobifuranones A, B and C were first isolated from the ascomycetes Gelasinospora kobi by Fujimoto et al. (1998). Kobiin posses a bicyclic skeleton of five- and fifteen-membered rings. Kobifuranones A, B and C were supposed to be metabolites formed from a common intermediate biosynthesized through the acetate-malonate pathway. AcOEt extracts of fungal mycelia containing kobiin and the three kobifuranones were found to suppress proliferation of mouse spleen lymphocytes stimulated with the mitogens CoA 180 Hesham El Enshasy and LPS. After solvent fractionation followed by repeated chromatography, the purified kobiin prepartion obtained showed the highest immunosuppressive activity. 7. Mycestericins Mycestericins are a group of unique immunosuppressive compounds and chemically, hydroxylated a-hydroxymethyl a-aminoalkanoic acids. All known types of mycestericins were isolated from the cultures of mycelia sterila (i.e. filamentous fungi without any morphological structures, neither sexual organs and spores nor conidia and other asexual spores). The chemical structures of mycestericin A (CAS 128440-98-2), B (CAS 128341-87-7), C (CAS 37817-99-5), D (CAS 157183-67-0) and E were determined on the basis of comprehensive spectroscopic studies and chemical tests (Sasaki et al. 1994). Mycestericins suppress the proliferation of lymphocytes in the mouse allogeneic mixed lymphocyte reaction with a potency similar to that of myriocin. Further studies led to the isolation of two more active compounds, mycestericins F and G, from the same fungus (Fujita et al. 1996). The chemical structures of mycestericins F and G were identical to the respective dihydromycestericins D and E. Mycestericin A has also been chemically synthesized using simple tartrate as starting compound (Sato et al. 2008); total chemical synthesis of mycestericins E was accomplished by a cinchona alkaloid-catalyzed asymmetric Baylis-Hillman reaction (Iwabuchi et al. 2001). Mycestericins D–G can enzymatically be prepared using L-threonine aldolase from Candida humicola in the key step reaction (Nishide et al. 2000). 8. Terprenins Terprenin (CAS 197899-11-9) was discovered by Kamigauchi et al. (1998) in the fermentation broth of Aspergillus candidus during a screening for natural immunosuppressants (Kamigauchi et al. 1998). It has a novel highly oxygenated p-terphenul structure with a prenyloxy side chain. Two terprenin derivatives, 3-methoxy-terprenin and 40 -deoxyterprenin, were also isolated from the fermentation liquid and showed significant immunosuppressive effects when tested with respect to the proliferation of mouse spleen lymphocytes. The most relevant activity of terprenin is its suppressive effect on the production of immunoglobulin E (IgE), which is a factor of 104 stronger than that of FK506, and interestingly, without any toxicological side-effect (Kawada et al. 1998). In mice experiments, terprenin suppressed IgE production in a typical dose dependent manner. Even after immunization with ovalbumin, when the IgE value had reached a high level, terprenin still exhibited a significant suppressive effect at 20–40 mg kg–1 (Liu 2006). The total synthesis of terprenin is possible and was reported by different authors (Kawada et al. 1998; Yonezawa et al. 1998). 9. FR901483 A potent immunosuppressant, FR901483, was isolated in 1996 from the fermentation broth of Cladobotryum sp. by Fujisawa Pharmaceutical Co. in Japan (Sakamoto et al. 1996). It was found to exert a potent immunosuppressive activity in vitro and significantly prolonged graft survival in the rat-skin allograft model, apparently by the inhibition of purine nucleotide biosynthesis. This compound has an intriguing tricyclic structure possessing a phosphate ester in its molecule. Since its discovery, FR901483 has garnered significant attention from the organic chemists due to its biological activity and unique aza-tricyclic nature. Thus, different synthesis schemes were published for the total synthesis of this important immunosuppressant (Maeng and Funk 2001; Kropf et al. 2006; Carson and Kerr 2009). 10. Colutellin A Colutellin A is a new immunosuppressive peptide recently isolated from Colletrichum dematium. It showed CDþ T-cell activation of interleukin 2 (IL-2) production with an IC50 of 167 nM. Moreover, it exhibited no cytotoxicity to human peripheral blood mononuclear cells in respective in vitro tests. Thus, it could be medicinally used as a novel immunosuppressive compound in the near future (Ren et al. 2008). III. Mushroom Immunomodulators Nowadays, immunostimulators (biological response modifiers) are becoming increasingly more popular in the health and wellness industries Immunomodulators as people have started to realize the importance of a healthy immune system as a first barrier for the prevention of diseases. These pro-drugs or prophylactic medicines have a long history in traditional medicine, in particular in Asian and Mediterranean countries. Thus the medical use of mushrooms has a long tradition in Japan, China, Korea and Southeast Asia, whereas, in Europe and the United States, this field has just been developing since the early 1980s. The positive medicinal properties of mushrooms are based on various cellular compounds and secondary metabolites, which can be isolated from different parts of the fruiting body or from the mycelium during growth in solid-state or liquid cultures (Tang et al. 2007). The immunomodulating effects of mushroom metabolites are especially valuable in the prophylaxis as a mild and non-invasive form of a treatment, which can even prevent the proliferation of metastatic tumors, and is used as a co-treatment in combination with classic chemo- and radiotherapies (Wasser 2002). The most potent immunomodulators produced by mushrooms belong to the lectins, terpenoids and polysaccharides. A. Lectins The immunomodulatory activities of lectins (highly glycolylated proteins with specific binding capacities) from different organisms have been known for decades. Mushroom lectins are characterized by their particular antiproliferative and antitumor activities. Boletus satanas lectin, bolesatine, was shown to have a potent mitogenic activity on human peripheral blood lymphocytes, and also to stimulate the release of IL-1a, IL2 and TNF-a from mononuclear cell cultures (Licastro et al. 1993). A fruiting- body lectin of Grifola frondosa showed cytotoxic activity against HeLa cells, when applied at low concentration (Kawagishi et al. 1990). A heterodimeric melibiose-binding lectin from fruiting bodies of the oyster mushroom Pleurotus ostreatus was reported to be an in vivo inhibitor of sarcoma S-180 and hepatoma H-22 tumor cells (Wang et al. 2000). A specific lectin was identified in fruiting bodies and mycelia of the straw mushroom (Volvariella volvacea) and had a stronger immunomodulatory effect than concanavalin A (She et al. 1998). Two lectins, TML-1 and TML-2, with immunomodulatory and antitumor activities were isolated from Tricholoma mongolicum; however, when these 181 lectins were directly tested in vitro, no antitumor activity was observed. This suggests that the lectins are rather immunomodulatory substances than substances exerting acute cytotoxicity. Peritoneal macrophages in mice treated with TML-1 or TML-2 revealed – after LPS stimulation – an enhanced production of nitrite and TNF-a. Both compounds inhibited also the growth of P815 mastocytoma cells by stimulating peritoneal macrophages to produce more macrophage-activating factors including interferon-g and some other cytokines (Wang et al. 1996, 1997). B. Terpenoids Terpenes are built up of isoprene sub-units consisting of five carbon atoms. Among the huge number of terpenes, are special triterpenoids which are exclusively found in certain macrofungi (mostly basidiomycetes) and are famous for their biological activities and medicinal properties. One example of such a triterpenoid compound is the highly oxidized lanostane which can be isolated from wood-decay fungi of the families Polyporaceae and Ganodermaceae (e.g. Ganoderma lucidum). This and related compounds show different biological activities including anti-infective, cytotoxic and immunomodulating efficacy (Moradali et al. 2007). Ganoderic, ganoderenic, ganodermic and applanoxidic acids, ganoderals, ganoderols, lucidone, ganodermanontriol as well as ganodermanondiol are the most common triterpenoids found in these mushrooms. Mixtures of these compounds can be prepared by the extraction of respective fruiting bodies with organic solvents. They were shown to have an antitumor activity that is comparable to that of certain b-D-glucans (see below). Fungal terpenoids can stimulate the NF-kB pathway and modulate Ras/Erk, c-myc and CREB proteins as well as mitogen-activated protein kinases (Gao et al. 2003). In consequence, these activation mechanisms can lead to other immune stimulations which are finally effective against tumor cells. C. Polysaccharides Various polysaccharides of microbial and nonmicrobial origin have been widely used as potential biological response modifiers (BRMs) as reviewed by Leung et al. (2006). Mushroom derived polysaccharides are regarded as excellent 182 Hesham El Enshasy immunostimulators due to their suitable therapeutic properties, i.e. they are barely toxic and have just negligible side-effects compared to other immunostimulants. Respective polysaccharides occur in relevant amounts in the macroscopic fruiting bodies and cultured mycelia but also to some extent in the culture filtrates of fungi. Most macrofungal polysaccharides belong either to the homoglycans or heteroglycans, and can bind to structure proteins to form polysaccharide-protein complexes. In general, immunomodulator polysaccharides appear to be related to the fungal cell wall and comprise (1!3)- and (1!6)-b-glucans as well as (1!3)-a-glucans or polysaccharide complexes of the galactomannan- and glucuromannan-protein type. Research on mushroom polysaccharides can be traced back to the 1960s when Ikegawa’s group in Japan first investigated the host-mediated antitumor activity of hot-water extracts of several edible mushrooms against sarcoma 180 cells of mice (Ikekawa et al. 1969). Until the late 1980s, three antitumor-immunomodulators of the b-glucan type were isolated and characterized, namely lentinan, schizophyllan and a protein-bound bglucan (PSK Kresin). They originate from the white-rot fungi Lentinus edodes, Schizophyllum commune and Coriolus versicolor, respectively, and have successfully been introduced into the probiotic and pharmaceutical market in Japan. A similar polysaccharopeptide as PSK, abbreviated as PSP is produced in China and widely used in the clinical treatment of tumors and in anticancer therapy (Ooi and Liu 2000). Although their mode of action against tumore cells is not yet fully understood, they have been demonstrated to act as biological response modifiers (BRMs), which are able to restore or enhance various immune responses in vitro and in vivo. The mushroom polysaccharides or polysaccharide-protein complexes stimulate the non-specific immune system and thereby exert antitumor activities through the stimulation of the body’s own defence mechanisms (Wasser and Weis 1999; Reshetnikov et al. 2001). They can activate effector cells like macrophages and T-lymphocytes or prompt NK cells to secrete cytokines like TNF-a, IFN-g and IL-1b. In turn, some of these cytokines are able to directly promote the cytotoxicity of macrophages. The production of cytokines by immune cells can be considered as a key event in the initiation and regulation of the body’s immune response (Lull et al. 2005). In this context, mushroom polysaccharides can act as antiproliferative effectors and induce apoptosis in tumor cells. Certain mushroom polysaccharides (e.g. BRMs) were shown to reduce the tumor size by more than 50% and considerably prolonged the survival of tumor-bearing mice (Wasser 2002). Though the exact mechanism of BRM action is not known, it has been proposed that they initiate a cascade of singal transduction that is responsible for the immune response. Since polysaccharides are not able to penetrate cells (due to their high molecular mass), the first step of the cascade may be the recognition of BRM and its specific binding to immunocell receptors. Some evidence exists that there are pattern recognition receptors (PRRs) for the molecular reception of the polysaccharide BRM (Lowe et al. 2001). The binding of the BRM-ligand to PRRs may initiate Rel/NF-kB-mediated signaling events, which leads to the induction of gene expression and the stimulation of specific cellular functions of the innate immunity system (Leung et al. 2006). Whilst it is known that mushroom extracts have immunomodulatory activity, the standard approach has always been to isolate, characterize and administer pure active compounds. However, different types of polysaccharides in a mushroom extract may have synergistic acitivities (Borchers et al. 2004; Lull et al. 2005). The responses to different polysaccharides are mediated by different PRRs on the cell surface. An appropriate combination of strong responses involving different parts of the cell may provide greater therapeutic effects than a single polysaccharide. A brief list of immunomodulator polysaccharides and polysaccharide–protein complexes from mushroom is given in Table 8.1. Mushroom polysaccharides greatly differ in their sugar composition, branching configuration, helical conformation and other physical properties. The structure relationship between immunomodulator and the anticancer activities of polysaccharides have been reviewed by several authors (Ooi and Liu 2000; Lull et al. 2005; Zhang et al. 2007; Ooi 2008). It has been stated that structural features such as (1!3)-b-linkages in the main chain of the glucan and additional (1!6)-b-branching points, represent important factors influencing their biological effectiveness. b-Glucans containing mainly (1!6)-linkages are less effective, maybe due to their inherent flexibility and the large number of possible conformations (Zhang et al. 2007). In general, b-glucans exhibit immunomodulatory and/or antitumor activities when their main chain (“backbone”) forms a linear structure and do not have long branches. For example, pachyman, a branched (1!3)b-D-glucan obtained from the brown-rot fungus Poria cocos is inactive, whereas pachymaran obtained by the debranching of pachyman using selective periodate oxidation and mild hydrolysis, shows a pronounced activity (Chihara et al. 1970a). Lentinan (2/5) and schizophyllan (1/3) are (1!3)-b-D glucans with two or one branches for every 5-D-glucopyranosyl and 3-D-glucopyranosyl residue, respectively (Tabata et al. 1981; Chihara 1992). The polysaccharide moiety in PSK (1/5) is a (1!3)-b-/(1!4)-b-D-glucan with one branch for every 5-D-glucopyranosyl residue (Tsukagoshi et al. 1984). Although the degree of their branches is different, their bioactivities are similar (Ooi and Liu 2000). Interstingly, debranched lentinan preparations were found to be more effective against cancer cells than the native lentinan when applied during in vivo studies Immunomodulators 183 Table 8.1. Immunomodulator polysaccharides from mushrooms Mushroom species Polysaccharide source Active component References Coriolus versicolor Mycelium, Culture broth Fruiting body, Mycelium, Culture broth Fruiting body, Mycelium Fruit body, Culture broth Polysaccharopeptide Cui and Chisti 2003 Glucan, heteroglycan, Cordyglucan Yalin et al. 2005 Russell and Paterson 2008 Glucan-protein complex, glycoprotein Heteroglycan, mannoglucan, glycopeptide Leung et al. 1997 Cordyceps sinensis. Flammulina velutipes Ganoderma lucidum Grifola frondosa Fruiting body, Culture broth Hericum erinaceus Fruiting body, Mycelium Fruiting body, Mycelium Fruiting body, Culture broth Inonotus obliquus Lentinus edodes Peziza verculosa Pleurotus ostreatus Sclerotinia sclerotiorum Fruiting body Fruit body, Mycelium, Culture broth Mycelium Fruiting body, Mycelium, Culture broth Sclerotium Tremella aurantialba Tricholoma lobayense Fruiting body Culture broth Polyporus umbellatus Polystrictus versicolar (Sasaki et al. 1976). Overall, the relationship between the molecule’s biological activity and the branching pattern/ ratio of b-glucans seems to be rather complicate. The available data indicate that the (1!3)-b-D-glucan backbone is essential and that the most active polymers have degrees of branching between 0.2 and 0.33 (Ooi 2008). The molecular weight of the polysaccharide plays also an important role for the bioactivity. For a (1!3)-b-glucan extract of G. frondosa consisting of fractions of different size, the highest immunomodulatory activity was detected for molecular masses around 800 kDa (Adachi et al. 1990). When PSK was separated into four fractions (F1, <50 kDa; F2, 50–100 kDa; F3, 100–200 kDa, F4, >200 kDa) by successive ultrafiltration, the highest immunomodulatory activity was obtained with the high-molecular mass fraction F4 (Kim et al. 1990). Chemically modified (1!3)-b-D-glucans, such as schizophyllan and lentinan having a linear “worm-like”, triple-helical structure and average molecular masses of <50 000 g mol–1 or >110 000 g mol–1 efficiently stimulated monocytes in vivo and caused the secretion of more TNF-a than the samples with molecular masses between 67 000 and 110 000 g mol–1 did Therefore, the actual Proteoglycan, glucan, heteroglycan, galactomannan, grifolan Heteroglycan, Heteroglycan-peptide Glucan Mannoglucan, glucan, Lentinan, polysaccharide protein complex Glucan, Proteoglycan Heteroglactan, Proteoglycan Glucan Heteroglycan, glycopeptides, krestin (PSK) Glucan, scleroglucan (SSG) Heteroglycan Polysaccharideprotein complex Miyazaki and Nishijima 1981 Gao and Zhou 2003 Cun et al. 1994 Yang et al. 2007 Mizuno 1992 Kim et al. 2005b Chihara et al. 1970b Hobbs 2000 Mimura et al. 1985 Sarangi et al. 2006 Yang et al. 2004 Cui and Chisti 2003 Palleschi et al. 2005 Survase et al. 2007 Liu et al. 2003 Liu et al. 1996 relation between the polysaccharide molecular mass and its immunomodulation activity remains to be clarified. Conformations of polysaccharides include single helices and triplehelices as well as random-coiled structures. A triple-helix conformation is usually more stable than a single-helix. Lentinan, schizophyllan and the glucan moiety of PSK have all triple helix structures. Also the cytokine-stimulating activity of (1!3)-b-D-glucans was found to be associated with the triple-helix conformation (Falch et al. 2000). Therefore, the immunological activities of polysaccharides must be dependent on appropriate helical conformation. To improve the biological activity of polysaccharides by chemical modification, carboxymethylated, hydroxylated, formylmethylated, aminethylated and sulfated products have been designed. For example, a hydroxylated schizophyllan was found to induce in vivo the production of higher concentrations of nitric oxide (NO) and TNF-a in macrophages than native 184 Hesham El Enshasy schizophyllan (Ohno et al. 1995). A sulfated (1!3)-a-D-glucan prepared by respective modification of native cell-wall glucan from L. edodes exhibited a strong antiproliferation activity against breast carcinoma cells, whereas its native water-insoluble precursor glucan had only moderate antitumor activity (Zhang and Cheung, 2002). Another study conducted by Zhang et al. (2004) showed that the high bioactivity of carboxymethylated b-glucan was mainly attributed to the increase of water-solubility and the latter was also the main factor that enhanced the efficacy of the hyperbranched b-glucan TM3b after sulfatation (Tao et al. 2006). D. Fungal Immunomodulator Proteins Recently, different mushrooms have been reported to produce a new family of fungal immunomodulatory proteins (FIPs) with possible applications in therapy (Chen and Wang 2007). These include Ling Zhi-8 (LZ-8) from Ganoderma lucidum (Kino et al. 1989), FIP-fve from Flammulina velutipes (Gr.) Sing (Ko et al. 1995), FIP-vvo and FIP-vvl from Volvariella volvacea (Bull.; Fr.) sing (Hsu et al. 1997), FIP-gts from Ganoderma tsugae Murr (Lin et al. 1997) and PCP from Poria cocos (Schw.) Wolf (Chang and Sheu 2007). All these compounds were grouped together in a distinct protein family based on similarities in their amino acids sequence and their effects on compounds of the immunological response system (Ko et al. 1995). FIPs were found to be mitogenic in vitro for human peripheral blood lymphocytes (hPBLs) and mouse splenocytes. They induce a bell-shaped dose–response curve similar to that of lectin mitogens. In the course of in vivo studies, the FIP-like substance LZ-8 could act as an immunosuppressive agent through the prevention of systemic anaphylactic reactions and significantly decreased footpad edema during the Arthus reaction (Tanaka et al. 1989). Moreover, it suppressed autoimmune diabetic reactions in diabetic mice and increased graft survival in transplanted allogenic mouse skin and pancreatic rats without producing the severe toxic effects known for CyA (Van der Hem et al. 1994; 1996). FIP-fve isolated from fruiting bodies of F. velutipes stimulated mitogenesis of human peripheral lymphocytes, suppressed systemic anaphylaxis reactions and enhanced the transcription of interleukin-2 (IL-2) and interferon-g (Ko et al. 1995). The induction mechanism of interferon-g production was proposed to be mediated by a signaling pathway involving the p38 mitogen-activated protein kinase (Wang et al. 2004). FIP-gts was reported to significantly induce cytokine secretion, cellular proliferation in human peripheral mononuclear cells (HPBMCs) and interferon-g expression. The effect of FIP-gts may be caused by the activation of phosphatidylinositol 3-kinase (Hsiao et al. 2008). Finally, the immunostimulus initiated by the recently isolated FIP-PCP is mediated via an enhanced production of NO, IL-1b, IL-6, IL-18 and TNF-a (Chang and Sheu 2007). E. Industrial Production of Mushroom Immunomodulators Immunomdulator metabolites can be isolated from fruiting bodies, cultured mycelia or culture filtrates. All medicinal mushrooms are lignocellulose degraders (white-rots, brown-rots, litter decomposers) and can utilize woody materials as growth substrates and for fruiting body production. One historical method of cultivation, that is still practiced mainly in Asia, is fungal cultivation on hardwood tree-logs. This process occurs over several years and yields two crops of mushrooms each year. It continues until the log physically “disappears” due to wood decay and lignocellulose decomposition. The use of polypropylene bags containing crushed lignocellulosics (including waste materials) and selected nutrients can be regarded as a modified version of the log method and actually represents a kind of solid-state fermentation (SSF). After autoclaving, the bags are inocultated with the mushroom mycelium of choice and can be incubated in the greenhouse under controlled conditions. This way, the production cycle for fruiting bodies can be shortened to 1–3 months (Smith et al. 2002). Mushroom production using SSF techniques was recently reviewed by Fan et al. (2008; see also Chapter 4 of this book). However, for the production of biomolecules, the production process should be carried out under more defined and controlled conditions to fulfil the strict requirements of the current good manufacturing practice (cGMP) for the production of active pharmaceutical ingredients (API). Quality control of mushroom cultivation poses several challenges, such as maintaining a constant substrate quality, temperature, moisture, a stable yield of the desired compounds and sterility. To overcome these problems, more specific research was carried to cultivate mushrooms under submerged conditions. This method of cultivation has some advantages over SSF, for example, high yields in fungal mycelium under more defined conditions in a closed and well controlled Immunomodulators volume (higher space–time yields). Furthermore, sterility is easier to guarantee in a stirred-tank bioreactor than in logs or plastic bags (Lull et al. 2005). Nowadays, different bioactive metabolites from mushrooms can successfully be produced in submerged culture both in form of intracellular and extracellular products. However, in order to scaleup these methods to an industrial scale, various technical problems will have to be solved (Tang et al. 2007). Like in case of other fungal metabolites, medium composition governs the bioactive agents’ production; for example, the production of mushroom polysaccharides was found to be regulated by the type and concentration of carbon and nitrogen souces (Wasser et al. 2003; Cui et al. 2006), the C/N ratio (Wang et al. 2005), by the ammonium ion concentration (Mao and Zhong 2006) and by different other components supplemented to the medium (Lim and Yun 2006). Other main key parameters influencing product yield are shear stress (Gong and Zhong 2005) and the mode of oxygen sypply to the culture (Tang and Zhong 2003). The steady-state concentration of the latter in the culture medium was found to be very important for the over-production of mushroom metabolites in bioreactors (Mao and Zhong 2004). Among different cultivation strategies applied, fed-batch cultivation was proved to be the method of choice for most mushrooms tested so far (Kim et al. 2006; Zou 2006). The recent reviews of Zhong and Tang (2004) and Tang et al. (2007) summarize and discuss the latest developments in this field. 185 are known so far. Assuming that the proportion of “useful” mushrooms among the undiscovered and unexamined species will be only 5%, this implies a number of 7000 promising species as a future source of immunomodulators (Lindequist et al. 2005). Moreover, it has to been taken into consideration that different strains of one species can produce different bioactive compounds. For example, strains of G. lucidum can produce more than 120 different triterpenes and in addition also bioactive polysaccharides and proteins. Based on the increased knowledge of the biochemistry and molecular biology of bioactive metabolites as well as the advanced technology of high-throughput screenings using omic approaches (genomics, proteomics, metabolomics), a rapid development of this research field is expected and will lead to the discovery of many novel immunomodulators in the near future. Not least, the investigation of novel immunomodulators and their effects will widen our knowledge of the complex mechanisms regulating the immune system and body defense. References IV. Conclusions Fungal immunomodulators act either as suppressors or stimulators, and they are equally important molecules for many medical applications. The number of known fungal immunomodulators is increasing every year through the discovery of new compounds from so-far ignored fungal species and the development of appropriate semi-synthetic derivatives. This has led to a rapid expansion of the respective market. Some of these agents have approved from FDA and are widely used as commercial preparations, some have reached the level of clinical trials and others are currently subject of detailed pre-clinical tests. Also in the future, the search for new safe immunomodulators (without severe side-effects) to substitute the currently used preparations will be an important goal for both the scientific and the industrial communities related to the health care sector. Mushrooms are one of the most promising sources for the production of safe and effective immunomdulators. The number of mushroom species (“macrofungi”) on earth has been estimated to be 140 000, suggesting that only 10% Aarnio TH, Agathos SN (1990) Pigmented variants of Tolypocladium inflatum in relation to cyclosporine A production. Appl Microbiol Biotechnol 33:435–437 Abdel Fattah YR, El Enshasy H, Anwar M, Omar H, Abou El Magd E, Abou Zahra R (2007) Application of factorial experimental designs for optimization of cyclosporine A production by Tolypocladium inflatum in submerged culture. J Microbiol Biotechnol 17: 1930–1936 Abraham EP (1945) The effect of mycophenolic acid on the growth of Staphylococcus aureus in heart broth. Biochem J 39:398–404 Adachi K, Chiba K (2007) FTY720 story. Its discovery and the following accelerated development of sphingosine 1-phosphate receptor agonists as immunomodulators based on reverse pharmacology. Perspect Med Chem 1:11–23 Adachi K, Kohara T, Nakao N, Arita M, Chiba K, Mishina T, Sasaki S, Fujita T (1995) Design, synthesis, and structure–activity relationships of 2-substituted-2amino-1,3-propanediols: discovery of a novel immunosuppressant, FTY720. Med Chem Lett 5:853–856 Adachi Y, Ohno N, Ohsawa M, Oikawa S, Yadomae T (1990) Change of biological activities of (1!3)-b-Dglucan from Grifola frondosa upon molecular-weight reduction by heat-treatment. Chem Pharmaceut Bull 38:477–481 Agathos SN, Lee J (1993) Mathematical modeling of the production of cyclosporine A by Tolypocladium inflatum: Effect of L-valine. Biotechnol Prog 9:54–63 186 Hesham El Enshasy Agathos SN, Marshall JW, Moraiti C, Parekh R, Madhosingh C (1986) Physiological and genetic factors for process development of cyclosporine fermentations. J Ind Microbiol 1:39–48 Agawal SS, Singh VK (1999) Immunomodulators: A Review of Studies on Indian Medicinal Plants and Synthetic Peptides. Proc Ind Natl Sci Acad B65:179–204 Alani F, Grove J, Anderson WA, Moo-Young M (2009) Mycophenolic acid production in solid-state fermentation using a packed-bed bioreactor. Biochem Eng J 44:105–110 Allison AC, Eugui EM (2000) Mycophenolate mofetil and its mechanisms of action. Immunopharmacology 47:85–118 Alloway RR (1999) Generic immunosuppressant use in solid organ transplantation. Transplant Proc 31 [Suppl 3A]:2S–5S Alsberg CL, Black OF (1913) Contribution of the study of maize deterioration; biochemical and toxicological investigations of Penicillium puberulum and Penicillium stolonigerum. Bull Burl Anim Ind US Dept Agric 270:1–47 Amano S, Ogawa N, Ohtsuka M, Ogawa S, Chida N (1998) Total synthesis and absolute configuration of FR65814. Chem Commun 1263–1264 Amano S, Ogawa N, Ohtsuka M, Chida N (1999) Chiral and stereoselective total synthesis of novel immunosuppressant FR65814 from D-glucose. Tetrahedron 55:2205–2224 Anderson WK, Boehm TL, Makara GM, Swann TR (1996) Synthesis and modeling studies with monocyclic analogues of mycophenolic acid. J Med Chem 39:46–55 Ando K, Suzuki S, Tamaura G, Arima K (1968) Antiviral activity of mycophenolic acid. Studies on antiviral and antitumor antibiotic IV. J Antibiot 21:649–652 Appel GB, Radhakrishnan J, Ginzler EM (2005) Use of mycophenolate mofetil in autoimmune and renal diseases. Transplantation 80[Suppl]: S265–S271 Aw MM, Verma A, Rela M, Heaton N, Mieli-Vergani G, Dhawan A (2008) Long-term outcome of mycophenolate rescue therapy for resistant acute allograft rejection in pediatric liver transplant recipients. Liver Transplant 14:1303–1308 Balakrishnan K, Pandey A (1996) Influence of amino acids on the biosynthesis of cyclosporine A by Tolypocladium inflatum. Appl Microbiol Biotechnol 45: 800–803 Banfi L, Beretta G, Colombo L, Gennari C, Scolastico C (1982) Total synthesis of (þ)-thermozymocidin (myriocin) from D-fructose. J Chem Soc Chem Commun 488–490 Bartman CD, Doerfler DL, Bird BA, Remaley AT, Peace JN, Campbell IM (1981) Mycophenolic acid production by Penicillium brevicompactum on solid media. Appl Env Microbiol 41:729–736 Bedford CT, Knittel P, Money T, Phillips GT, Salisbury P (1973) Biosynthesis of mycophenolic acid. Can J Chem 51:694–697 Benedetti A, Colombo E, Nichele M, Pagani H (2002) Process for producing mizoribine by Eupenicillium javanicum. Patent EP 19990103099 Bentley R (2000) Mycophenolic acid: a one hundred year odyssey from antibiotic to immunosuppressant. Chem Rev 100:3801–3825 Birch AJ, English RJ, Massy-Westropp RA, Smith H (1958) Studies in relation to biosynthesis. Part XIV. The origin of the nuclear methyl groups in mycophenolic acid. J Chem Soc 1958:365–368 Birkinshaw JH, Raistrick H, Ross DJ (1952) The molecular constitution of mycophenolic acid, A metabolic product of Penicilliusm brevicompactum Dierckx. Part 3. Further observations on the structural formula for mycophenolic acid. Biochem J 50:630–634 Bissett J (1983) Notes on Tolypocladium and related genera. Can J Bot 61:1311–1329 Boiteau J-G, Van de Weghe P, Eustache J (2001) A new, ring closing metathesis-based synthesis of (–)-fumagillol. Org Lett 3:2737–2740 Borchers AT, Keen CL, Gerhwin ME (2004) Mushrooms, tumors, and immunity: an update. Exp Biol Med 229:393–406 Budde K, Giessing M, Liefeldt L, Neumayer H-H, Glander P (2006) Moderne immunsuppressiva nach Nierentransplantation. Standard oder maßgeschneidert? Urologe 45:9–17 Butch AW (2008) Immunosuppressive drugs. In: Dasgupta A (ed) Handbook of drug monitoring methods. Humana, Totowa, pp 165–199 Carson CA, Kerr MA (2009) Total synthesis of FR901483. Org Lett 11:777–779 Carter SB, Franklin TJ, Jones DF, Leonard BJ, Mills SD, Turner RW, Turner WB (1969) Mycophenolic acid: An anti-cancer compound with unusual properties. Nature 223:848–850 Cattran DC, Wang MM, Appel G, Matalon A, Briggs W (2004) Mycophenolate mofetil in the treatment of focal segmental glomerulosclerosis. Clin Nephrol 62:405–411 Chang H-H, Sheu F (2007) A novel fungal immunomodulatory protein (PCP) isolated from Poria cocos activates mouse peritoneal macrophage involved in tolllike receptor 4. FASEB J 21:702.15 Chen L, Gao G, Felczak K, Bonnac L, Patterson SE, Wilson D, Bennett EM, Jayaram HN, Hedstrom L, Pankiewicz KW (2007) Probing binding requirements of type I and type II isoforms of inosine monophosphate dehydrogenase with adenine-modified nicotinamide adenine dinucleotide analogues. J Med Chem 50:5743–5751 Chen L, Wilson DJ, Labello NP, Jayaram HN, Pankiewicz KW (2008) Mycophenolic acid analoges with a modified metabolic profile. Bioorg Med Chem 16: 9340–9345 Chen T-C, Wang T-S (2007) Use of fungal immunomodulatory protein. Patent EP 20060017781 Chihara G (1992) Immunopharmacology of lentinan, a polysaccharide isolated from Lentinus edodes: Its application as a host defense potentiator. Int J Orient Med 17:57–77 Chihara G, Hamuro J, Maeda Y, Arai Y, Fukuoka F (1970a) Antitumour polysaccharide derived chemically from natural glucan (pachyman). Nature 225: 934–944 Immunomodulators Chihara G, Hamuro J, Maeda YY, Arai Y, Fukuoka F (1970b) Fractionation and purification of the polysaccharides with marked antitumor activity, especially lentinan, from Lentinus edodes. Cancer Res 30:2776–2782 Chun GT, Agathos SN (1989) Immobilization of Tolypocladium inflatum spores into porous celite beads for cyclosporine production. J Biotechnol 9:237–254 Cline JC, Nelson JD, Gerzon K, Williams RH, Delong DC (1969) In vitro antiviral activity of mycophenolic acid and its reversal by guanine compounds. Appl Microbiol 18:14–20 Clutterbuck PW, Oxford AE, Raistrick H, Smith G (1932) Studies in the biochemistry of micro-organisms: the metabolite products of the Penicillium brevicompactum series. Biochem J 26:1441–1485 Contreas M, DeSilva M (1994) The prevention and management of the hemolytic disease of newborn. J R Soc Med 87:256–258 Covarrubias-Zúñiga A, Gonzlez-Lucas A (1998) A total synthesis of mycophenolic acid. Tetrahedron Lett 39:2881–2882 Covarrubias-Zúñiga A, Gonzalez-Lucas A, Domı́nguez MM (2003) Total synthesis of mycophenolic acid. Tetrahedron 59:1989–1994 Cui FJ, Chisti Y (2003) Polysaccharopeptides of Coriolus versicolor: physiological activity, uses and production. Biotechnol Adv 21:109–122 Cui FJ, Li Y, Xu ZH, Sun K, Tao WY (2006) Optimization of the medium composition for production of mycelia biomass and exo-polymer by Grifola frondosa GF9801 using response surface methodology. Bioresour Technol 97:1209–1216 Cun Z, Mizuno T, Ito H, Shimura K, Sumiya T, Kawade M (1994) Antitumor activity and immunological property of polysaccharides from the mycelium of liquidcultures Grifola frondosa. J Jpn Soc Food Sci Technol 41:724–733 Dittmann J, Wenger RM, Kleinkauf H, Lawen A (1994) Mechanism of cyclosporine A biosynthesis. Evidence for biosynthesis via a single linear undecapeptide precursor. J Biol Chem 269:2841–2846 Doerfler DL, Nultron CP, Bartman CD, Gottlieb FJ, Campbell IM (1978) Spore germination, colony development, and secondary metabolism in Penicillium brevicompactum: a radiogas chromatographic and morphological study. Can J Microbiol 24:1490–1501 Doerfler DL, Bartman CD, Campbell IM (1979) Mycophenolic acid production by Penicillium brevicompacturm in two media. Can J Microbiol 25:940–943 Dreyfuss MM, Gams W (1994) Proposal to reject Pachybasium niveum Rostr. In order to retain the name Tolypocladium inflatum W. Gams for fungus that produces cyclosporin. Taxon 43:660–661 El-Araby ME, Bernacki RJ, Makara GM, Pera PJ, Anderson WK (2004) Synthesis, molecular modeling, and evaluation of nonphenolic indole analogs of mycophenolic acid. Bioorg Med Chem 12:2867–2879 El Enshasy H (2007) Filamentous fungal cultures – process characteristics, products, and applications. In: Yang ST (ed) Bioprocessing for value-added products from renewable resources. Elsevier, Rotterdam, pp 225–261 187 El Enshasy H, Abdel Fattah Y, Atta A, Anwar M, Omar H, Abou EL Magd S, Abou Zahra R (2008) Kinetics of cell growth and cyclosporine A production by Tolypocladium inflatum when scaling up from shake flask to bioreactor. J Microbiol Biotechnol 18:913–917 Engel G, von Milczewski EE, Prokopek D, Teuber M (1982) Strain-specific synthesis of mycophenolic acid by P. roqueforti in blue-veined cheese. Appl Env Microbiol 43:1034–1040 Epinette WW, Parker CM, Jones EI, Greist MC (1987) Mycophenolic acid for psoriasis. J Am Acad Dermatol 17:962–971 Eugui EM, Allison AC (1993) Immunosuppressive activity of mycophenolic mofetil. Ann NY Acad Sci 685:309–329 Falch BH, Espevik T, Ryan L, Stokke BT (2000) The cytokine stimulating activity of (1!3)-b-D-glucans is dependent on the triple helix conformation. Carbohydr Res 329:587–596 Fan L, Soccol CR, Pandey A (2008) Mushroom production. In: Pandey A, Soccol CR, Larroche C (eds) Current developments in solid-state fermentation. Springer ScienceþBusiness Media, New York, pp 253–274 Ferraresso M, Kahan BD (1993) New immunosuppressive agents for pediatric transplantation. Pediatr Nephrol 7:567–573 Figg WD, Pluda JM, Lush RM, Saville MW, Wyvill K, Reed E, Yarchoan R (1997) The pharmacokinetics of TNP-470, a new angiogenesis inhibitor. Pharmacotherapy 17:91–97 Foster BC, Coutts RT, Pasutto FM, Dossetor JB (1983) Production of cyclosporin A by carrageenan-immobilized Tolypocladium inflatum in an airlift reactor with external loop. Biotechnol Lett 5:693–696 Fuji K, Fujita E, Takaishi Y, Fujita T, Arita I, Komatsu M, Hirasuki N (1978) New antibiotics, trichopolyns A and B: isolation and biological activity. Experientia 34:237–239 Fujimoto H, Nakayama Y, Yamazaki M (1993) Identification of immunosuppressive components of musrhoom, Lactarius flavidulus. Chem Pharm Bull 41:654–658 Fujimoto H, Satoh Y, Yamazaki M (1998) Four new immunosuppressive components, kobiin and kobifuranones A, B, and C, from an ascomycetes, Gelasinospora kobi. Chem Pharm Bull 46:211–216 Fujino M, Li XK, Kitazawa Y, Guo L, Kawasaki M, Funeshima N, Amano T, Suzuki S (2002) Distinct pathways of apoptosis triggered by FTY720, etoposide, and anti-fas antibody in human T-lymphoma cell line (Jurkat cells). Pharmacol Exp Therap 300:939–945 Fujita T, Takaishi Y, Okamura A, Fujita E, Fuji K, Hirasuka N, Momatsu M, Arita I (1981) New peptide antibiotics, tricopolyns I and II from Trichoderma polysporum. J Chem Soc Chem Commun 585–587 Fujita T, Inoue K, Yamamoto S, Ikumoto T, Sasaki S, Toyama R, Chiba K, Hoshino Y, Kumoto T (1994) Fungal metabolites. Part 11. A potent immunosuppressive activity found in Isaria sinclairii metabolite. J Antibiot 47:208–215 Fujita T, Hamamichi N, Kiuchi M, Matsuzaki T, Kito Y, Inoue K, Hirose R, Yoneta M, Sasaki S, Chiba K (1996) Determination of absolute configuration and 188 Hesham El Enshasy biological activity of new immunosuppresants, mycestericins D, E, F and G. J Antibiot 49:846–853 Gams W (1971) Tolypocladium. Eine Hyphomycetengattung mit geschwollenen Philaliden. Persoonia 6:185–191 Gao Y, Zhou S (2003) Cancer prevention and treatment by Ganoderma, a mushroom with medicinal properties. Food Rev Int 19:275–325 Gao Y, Zhou S, Chen G, Dai X, Ye J (2003) A phase I/II study of a Ganoderma lucidum (Curt.: Fr.) P. Karst. Extract (ganopoly) in patients with advanced cancer. Int J Med Mushroom 4:207–214 Gong HG, Zhong JJ (2005) Hydrodynamic shear stress affects cell growth and metabolite production by medicinal mushroom Ganoderma lucidum. Chin J Chem Eng 13:426–428 Gregory CR, Kyles AE, Bernsteen L, Wagner GS, Tarantal AF, Christe KL, Brignolo L, Spinner AG, Griffey SM, Paniagua RT, Hubble RW, Borie DC, Morris RE (2004) Compared with cyclosporine, ISATX247 significantly prolongs renal-allograft survival in a nonhuman primate model. Transplantation 78:681–685 Gullo VP, McAlpine J, Lam KS, Baker W, Petersen F (2006) Drug discovery from natural products. J Ind Microbiol Biotechnol 33:523–531 Hartmann N, Enk A (2005) Mycophenolate mofetil and skin diseases. Lupus 14 [Suppl 1]:S58–S63 Hatanaka H, Kino T, Hashimoto M, Tsurumi Y, Kuroda A, Tanaka H, Goto T, Okuhara M (1988) FR65814, a novel immunosuppressant isolated from a Penicillium strain. J Antibiot 41:999–1008 Hayashi M, Kim YP, Takamatsu S, Preeprame S, Komiya T, Masuma R, Tanaka H, Komiyama K, Omura S (1996) Chlovalicin, a new cytocidal antibiotic produced by Sporothrix sp. I. Taxonomy, fermentation, isolation and biological activities. J Antibiot 49:631–634 Hiestand, PC, Gunn H, Gale J (1985) The immunosuppressive profile of a new natural cyclosporine analogue: Nva2-cyclosporine. Transplant Proc 17:1362–1364 Hiestand PC, Graber M, Hurtenbach V, Hermann P, Borel JF (1992) The new cyclosporine derivative, SDZ IMM 125: in vitro and in vivo pharmacological effects. Transplant Proc 24:31–38 Hobbs C (2000) Medicinal values of Lentinus edodes (Berk.) Sing. (Agaricomyvetideae). A literature review. Int J Med Mushrooms 2:287–297 Hodge KT, Krasnoff SB, Humber RA (1996) “Tolypocladium inflatum is the anamorph of Cordyceps subsessilis”. Mycologia 88:715–719 Hoffmann K, Schneider-Scherzer E, Klein-Kauf H, Zocher R (1994) Purification and characterization of an eukaryotic alanine racemas, acting as a key enzyme in cyclosporine biosynthesis. J Biol Chem 269:12710–12714 Hood KA, Zarembski DG (1997) Mycophenolate mofetil: a unique immunosuppressive agent. Am J Health Sys Pharm 54:285–294 Hoppert M, Gentzsch C, Schörgendorfer K (2001) Structure and localization of cyclosporine synthetase, the key enzyme of cyclosporine biosynthesis in Tolypocladium inflatum. Arch Microbiol 176:285–293 Hsiao Y-M, Huang Y-L, Tang S-C, Shieh G-J, Lai J-Y, Wang H, Ying T-H, Ko J-L (2008) Effect of a fungal immunomodulatory protein from Ganoderma tsugae on cell cycle and interferon-gamma production through phosphatidylinositol 3-kinase signal pathway. Process Biochem 43:423–430 Hsu H-C, Hsu C-I, Lin R-H, Kao C-L, Lin J-Y (1997) Fipvvo, a new fungal immunomodulatory protein isolated from Volvariella volvacea. Biochem J 323: 557–565 Iaccarino L, Rampudda M, Canova M, Della Libera S, Sarzi-Puttinic P, Doria A (2007) Mycophenolate mofetil: what is its place in the treatment of autoimmune rheumatic diseases? Autoimmun Rev 6:190–195 Iijima M, Masuda T, Nakamura H, Naganawa H, Kurasawa S, Okami Y, Ishizuka M, Takenchi T, Itaka Y (1992) Metacytofilin, a novel immunomodulator produced by Metarhizium sp. TA2759. J Antibiot 45:1553–1556 Ikekawa T, Uehara N, Maeda Y, Nakanish M, Fukuoka F (1969) Antitumor activity of aqueous extracts of edible mushrooms. Cancer Res 29:734–735 Im D-S (2003) Linking Chinese medicine and G-proteincoupled receptors. Trends Pharmacol Sci 24:2–4 Iwabuchi Y, Furukawa M, Esumi T, Hatakeyama S (2001) An enantio- and stereocontrolled synthesis of (–)mycestericin E via cimchona alkaloid-catalyzed asymmetric Baylis–Hillman reaction. Chem Comm (Comb) 19:2030–2031 Jaureguiberry G, Farragia-Fougerouse G, Audier H, Lederer E (1964) On transfer of the CD-3 group of methionine-CD-3 during biosynthesis of mycophenolic acid. CR Acad Sci Paris 259:3108–3110 Jekkel A, Barta I, Kónya A, Sütő J, Boros S, Horvth G, Ambrus G (2001) Microbiological transformation of mycophenolic acid. J Mol Cat B Enz 11:423–426 Jekkel A, Barta I, Boros S, Sütő J, Horvth G, Szabó Z, Ambrus G (2002) Microbiological transformation of mycophenolic acid. Part II. J Mol Cat B Enz 19/ 20:209–214 Jones MC, Marsden SP (2008) Total synthesis of the immunosuppressants myriocin and 2-epi-myriocin. Org Lett 10:4125–4128 Jorgensen KA, Koefoed-Nielsen PB, Karamperis N (2003) Clacineurin phosphate activity and immunosuppression. A review on the role of calcineurin phosphate activity and the immunosppressive effect of cyclosporin A and tacrolimus. Scand J Immunol 57:93–98 Kahan BD (1992) New immunosuppressive drugs: needs in an applications to pediatric transplantation. Eur J Pediatr 151[Suppl 1]:S9–S12 Kallen J, Mikol V, Quesniaux VFJ, Walkinshaw MD, Schneider-Scherzer E, Schörgendorfer K, Weber G, Fliri HG (1997) Cyclosporins: Recent developments in biosynthesis, pharmacology and biology, and clinical applications. In: Rehm, H-J. and Reed G (eds) Biotechnology, VCH, Weinheim, pp 535–591 Kamigauchi T, Sakazaki R, Nagashima K, Kawamura Y, Yasuda Y, Matsushima K, Tani H, Takahashi Y, Ishii K, Suzuki R, Koizumi K, Nakai H, Ikenishi Y, Terui Y (1998) Terprenin, novel immunosuppressants produced by Aspergillus candidus. J Antibiot 51:445–450 Immunomodulators Kawada K, Arimura K, Tsuri T, Fuji M, Komurasaki T, Yonezawa S, Kugimiya A, Haga N, Mitsumori S, Inagaki M, Nakatani T, Tamura Y, Takechi S, Taishi T, Kishino J, Ohtani M (1998) Total synthesis of terprenin, a highly potent and novel immunoglobulin E antibody suppressant. Ang Chem Int Ed 37:973–975 Kawagishi H, Nomura A, Mizuno T, Kimura A, Chiba S (1990) Isolation and characterization of lectin from Grifola frondosa fruiting bodies. Biochim Biophys Acta 1034:247–252 Khan MM (2008) Immunosuppressive agents. In: Khan MM (ed) Immunopharmacology Springer ScienceþBusiness Media, New York, pp 87–105 Khedkar AP, Subramaniyam P, Ananda K, Ramkrishna M, Tambe SP, Ganesh S, Sircar A, Shrikumar S (2007) Manufacture and purification of cyclosporine A. US Patent 7,176,001 Kida T, Takehiko I, Hiroshiro S (1984) Method for production of mycophenolic acid by fermentation. US Patent 4,452,891 Kim D, Ahn SK, Bae H, Choi WJ, Kim HS (1997) An asymmetric total synthesis of (–)-fumagillol. Tetrahedron Lett 38:4437–4440 Kim D, Ahn SK, Bae H, Kim HS (2005a) A stereoselective asymmetric synthesis of antibiotic (–)-fumagillol using claisen rearrangement and intramolecular ester enolate alkylation as key steps. Arch Pharm Res 28:129–141 Kim F, Sakagami H, Tanuma SI, Konno K (1990) Stimulation of interferon-g-induced human myelogenous leukemic cell differentiation by high molecular weight PSK subfraction. Anticancer Res 10:55–58 Kim HM, Paik SY, Ra KS, Koo KB, Yun JW, Choi JW (2006) Enhanced production of exopolysaccharides by fed-batch culture of Ganoderma resinaceum DG6556. J Microbiol 44:233–242 Kim YO, Han SB, Lee HW, Ahn HJ, Yoon YD, Jung JK, Kim HM, Shin CS (2005b) Immuno-stimulating effect of the endo-polysaccharide produced by submerged culture of Inonotus obliquus. Life Sci 77:2438–2456 Kino K, Yamashita A, Yamaoka K, Watanabe J, Tanaka S, Ko K, Shimizu K, Tsunoo H (1989) Isolation and characterization of a new immunomodulatory protein, Ling Zhi-8 (LZ-8), from Ganoderma lucidium. J Biol Chem 264:472–478 Klatt J, Hartung H-P, Hohlfeld R (2007) FTY720 (Fingolimod) als neue Therapiemöglichkeit der Multiplen Sklerose. Nervenarzt 78:1200–1208 Kluepfel D, Bagli J, Baker H, Charest M-P, Kudelski A, Sehgal SN, Vézina C (1972) Myriocin, a new antifungal antibiotic from Myriococcum albomyces. J Antibiot 25:109–115 Kluepfel D, Kudelski A, Bagli J (1975) Myriocin and process of preparation. US Patent 3,928,572 Ko JL, Hsu CI, Lin RH, Kao CL, Lin JY (1995) A new fungal immunomodulatory protein, FIP-fve isolated from the edible mushroom, Flammulina velutipes and its complete amino acid sequence. Eur J Biochem 228:244–249 Kratochvil B, Jegorov A, Pakhomova S, Husk M, Bulej P, Cvak L, Sedmera P, Havlicek V (1999) Crystal structure of cyclosporine derivatives: O-acetyl-(4R)-4- 189 (E-2-butyl)-4,N-dimethyl-L-threonyl-cyclosporin A and O-acetyl-(4R)-4-[E-2-(4-bromobutyl)]-4,Ndimethyl-L-threonyl-cyclosporin A. Collect Czec Chem Commun 64:89–98 Krishna C (2005) Solid-state fermentation systems – an overview. Crit Rev Biotechnol 25:1–30 Kroll M, Arenzana-Seisdedos F, Bachelerie F, Thomas D, Fringuet B, Conconi M (1999) The secondary fungal metabolite gliotoxin targets proteolytic activities of the proteasome. Chem Biol 6:689–698 Kropf JF, Meigh IC, Bebbington MWP, Weinreb SM (2006) Studies on a total synthesis of the microbial immunosuppressive agent FR901483. J Org Chem 71:2046–2055 Kuboki H, Tsuchida T, Wakazono K, Isshik K, Kumagai H, Yoshioka T (1999) Mer-f3, 12-hydroxy-ovalicin, produced by Metarhizium sp. F3. J Antibiot 52:590–593 Kupfahl C, Ruppert T, Dietz A, Geginat G, Hof H (2007) Candida species fail to produce the immunosuppressive secondary metabolite gliotoxin in vitro. FEMS Yeast Res 7:986–992 Lafont P, Debeaupuis JP, Gaillardin M, Payen J (1979) Production of mycophenolic acid by Penicillium roqueforti strains. Appl Env Microbiol 37:365–368 Lawen A, Zocher R (1990) Cyclosporin synthetase. The most complex peptide synthesizing multienzyme polypeptide so far described. J Biol Chem 256: 11355–11360 Lee J, Agathos SN (1989) Effect of amino acids on the production of cyclosporine A by Tolypocladium inflatum. Biotechnol Lett 11:77–82 Lee TH, Chun G-T, Chang YK (1997) Development of sporulation/immobilization method and its application for the continuous production of cyclosporin A by Tolypocladium inflatum. Biotechnol Prog 13:548–550 Lee WA, Gu L, Miksztal AR, Chu N, Leung K, Nelson PH (1990) Bioavailability improvement of mycophenolic acid through amino ester derivatization. Pharm Res 7:161–166 Leung MY, Fung KP, Choy YM (1997) The isolation and characterization of an immunomodulatory and antitumor polysaccharide preparation from Flammulina velutipers. Immunopharmacol 35:255–263 Leung MYK, Liu C, Koon JCM, Fung KP (2006) Polysaccharide biological response modifiers. Immunol Lett 105:101–114 Licastro F, Morini MC, Kretz O, Dirheimer G, Creppy EE, Stirpe F (1993) Mitogenic activity and immunological properties of bolesatine, a lectin isolated from the mushroom Boletus satanas Lenz. Int J Biochem 25:789–792 Lida A, Mihara T, Fujita T, Takaishi Y (1999) Peptidic immunosuppressants from the fungus Trichoderma polysporum. Bioorg Med Chem Lett 24:3393–3396 Lim JM, Yun JW (2006) Enhanced production of exopolysaccharides by supplementation of toluene in submerged culture of an edible mushroom Collybia maculate TG-1. Process Biochem 41:1620–1626 Lin WH, Hung CH, Hsu CI, Lin JY (1997) Dimerization of the N-terminal amphipathic alpha-helix domain of the fungal immunomodulatory protein from Ganoderma tsugae (Fip-gts) defined by yeast two hybrid 190 Hesham El Enshasy system and site-directed mutagenesis. J Biol Chem 272:20044–20048 Lindequist U, Niedermeyer THJ, Jülich W-D (2005) The pharmaceutical potential of mushrooms. eCAM 2:285–299 Liu C, Xie H, Su H, Han J, Liu Y (2003) Anti-thrombus effect on the fermented products of mycelium from Tremella aurantialba. Nat Prod Res Dev 3:35–37 Liu F, Ooi VEC, Liu WK, Chang ST (1996) Immunomodulation and antitumor activity of polysaccharide–protein complex from culture filtrate of a local edible mushroom, Tricholoma lobayense. Gen Pharmacol 27:621–624 Liu J-K (2006) Natural terphenyls: developments since 1877. Chem Rev 106:2209–2223 Liu S, Widom J, Kemp CW, Crews CM, Clardy J (1998) Structure of human methionine aminopeptidase-2 complexed with fumagillin. Science 282:1324–1327 Liu V, Mackool BT (2003) Mycophenolate in dermatology. J Dermatol Treat 14:203–211 Logothetis CJ, Wu KK, Finn LD, Daliani D, Figg W, Ghaddar H, Gutterman JU (2001) Phase I trial of the angiogenesis inhibitor TNP-470 for progressive androgen-independent prostate cancer. Clin Cancer Res 7:1198–1203 Loosli HR, Oschkinat H, Weber HP, Petcher T (1985) The conformation of cyclosporine A in the crystal and in solution. Helv Chim Acta 69:682–704 Lowe E, Rice P, Ha TZ, Li CF, Kelley J, Ensley H, Lopez-Perez J, Kalbfleisch J, Lowman D, Margl P, Browder W, Williams D (2001) A (1!3)-b-D-linked heptasaccharide is the unit ligand for glucose pattern recognition receptors on human monocytes. Microbes Infect 3:789–797 Lull C, Wichers HJ, Savelkoul HFJ (2005) Antiinflammatory and immunomodulating properties of fungal metabolites. Mediat Inflam 2:63–80 Ly M, Margaritis A (2007) Effect of temperature on the extraction kinetics and diffusivity of cyclosporine A in the fungus Tolypocladium inflatum. Biotechnol Bioeng 96:945–955 Ly M, Margaritis A, Jajuee B (2007) Effect of solvent concentration on the extraction kinetics and diffusivity of cyclosporine A in the fungus. Tolypocladium inflatum. Biotechnol Bioeng 96:67–79 Mao XB, Zhong JJ (2004) Hyperproduction of corydycepin by two-stage dissolved oxygen control in submerged cultivation of medicinal mushroom Cordyceps militaris in bioreactors. Biotechnol Prog 20:1408–1413 Mao XB, Zhong JJ (2006) Significant effect of NH4þ on cordycepin production by submerged cultivation of medicinal mushroom Cordyceps militaris. Enz Microb Technol 38:343–350 Marahiel MA, Stachelhaus T, Mootz HD (1997) Modular peptide synthetases involved in non-ribosomal synthesis. Chem Rev 97:2651–2673 Matsuzawa Y, Nakase T (1984) Metabolite fate of ethyl O[N-(p-carboxyphenyl)-carbamoyl] mycophenolate (CAM), a new antitumor agent, in experimental animals. J Pharmacobiodyn 7:776–783 Mazitschek R, Huwe A, Giannis A (2005) Synthesis and biological evaluation of novel fumagillin and ovalicin analogues. Org Biomol Chem 3:2150–2154 McCorkindale NJ Baxter RL (1981) 4-methyl- and 7methylphthalan-1-one derivatives of mycophenolic acid: use in establishing the distribution of acetate derived hydrogens. Tetrahedron 37:2131–2136 Menza-Aviña ME, Ordoñez M, Fernndez-Zertuche M, Rodriguez-Fragoso L, Esparza JR, de los Rı́os-Corsino AAM (2005) Synthesis of some monocyclic analogues of mycophenolic acid via the Johnson ortho ester Claisen rearrangement. Bioorg Med Chem 13:6521–6528 Mi-Jin L, Duong CTP, Han K, Kim E-S (2009) Combination strategy to increase cyclosporin A productivity by Tolypocladium niveum using random mutagenesis and protoplast transformation. J Microbiol Biotechnol 19:869–872. doi: 10.4014/jmb.0811.629 Mimura H, Ohno N, Suzuki I, Yadomae T (1985). Purification, antitumor activity, and structural characterization of b-1,3-glucan from Peziza versiculosa. Chem Pharm Bull 33:5096–5099 Mitsui H, Matsuno T, Ogawa H, Shiio T, Yugari Y, Tamura G (1981) Antitumor activity of a new compound, ethyl O-[N-(p-carboxyphenyl)-carbamoyl]mycophenolate, against various experimental tumors upon oral administration. Gann 72:66–71 Miyake Y, Kozutsumi Y, Nakamura S, Fujita T, Kawasaki T (1995) Serine palmitoyltransferase is the primary target of a sphingosine-like immunosuppressant, ISP-1/ myriocin. Biochem Biophys Res Commun 211:396–403 Miyazaki T, Nishijima M (1981) Studies on fungal polysaccharides. XXVII. Structural examination of a water-soluble, antitumor polysaccharide of Ganoderma lucidum. Chem Pharm Bull 29:3611–3625 Mizobuchi M, Iwasaki Y, Sako K, Kaneko Y (1997) Suppression of acute experimental allergic encephalomyelitis in Lewis rats with a mycophenolic acid derivative. Tohoku J Exp Med 182:217–229 Mizuno K, Tsujino M, Takada M, Hayashi M, Atsumi K, Asano K, Matsuda T (1974) Studies on bredinin, I. Isolation, characterization and biological properties. J Antibiot 27:775–782 Mizuno K, Takuji A, Masatoshi T, Masaki T, Munetoshi Y, Tetsuo M, Mitsuo H (1975) 4-Carbamoyl-1-b-D-ribofuranosylimidazolium-5-olate. US Patent 3,888,843 Mizuno T (1992) Antitumor active polysaccharides isolated from the fruiting body of Hericium erinaceum, and edible, and medicinal mushroom called Yamabushitake or Houtou. Biosci Biotechnol Biochem 56:349–357 Molino BF, Yang Z (2006) Novel cyclosporine analogues and their pharmaceutical uses. US Patent 2006/ 0069015 A1 Moradali M-F, Mostafavi H, Chods S, Hedjaroude G-A (2007) Immunomodulating and anticancer agents in the realm of macromycetes fungi (macrofungi). Int Immunopharmacol 7:701–724 Moussaı̈f M, Jacques P, Schaarwächter P, Budzikiewicz H, Thonart P (1997) Cyclosporin C is the main antifungal compound produced by Acremonium luzulae. Appl Environ Microbiol 63:1739–1743 Muth WL, Nash III CH (1975) Biosynthesis of mycophenolic acid: purification and characterization of S-adenosyl-Lmethionine: Demethylmycophenolic acid O-methyltransferase. Antimicrob Agents Chemother 8:321–327 Immunomodulators Mydlarski PR (2005) Mycophenolate mofetil: a dermatologic perspective. Skin Ther Lett 10:1–6 Nakajima S, Nozawa K (1979) Isolation in high yield citrinin from Penicillium odoratum and of mycophenolic acid from Penicillium brunneostoloniferum. J Nat Prod 42:423–426 Nakajima H, Hasasaki T, Tanaka K, Kimura Y, Udagawa S-I, Horie Y (1989) Production of cyclosporine by fungi belonging to the genus Neocosmospora. Agric Biol Chem 53:2291–2292 Nawata Y, Kuriki Y, Haneda M, Ochi K, Mori T (1988) Structure of a mycophenolic acid derivative (CAM). Acta Cryst C44:196–198 Nawata Y, Kuriki Y, Haneda M, Ochi K, Mori T (1989) Crystal structure of a mycophenolic acid derivative, CAM (form Ia). Anal Sci 5:119–120 Nelson PH, Eugul E, Wang CC, Allison AC (1990) Synthesis and immunosuppressive activity of some sidechain variants of mycophenolic acid. J Med Chem 33:833–838 Nelson PH, Carr SF, Devens BH, Eugui EM, Franco F, Gonzalez C, Hawley RC, Loughhead DG, Milan DJ, Papp E, Patterson JW, Rouhafza S, Sjogren EB, Smith DB, Stephenson RA, Talamas FX, Waltos A-M, Weikert RJ, Wu JC (1996) Structure–activity relationships for inhibition of inosine monophosphate dehydrogenase by nuclear variants of mycophenolic acid. J Med Chem 39:4181–4196 Niido O, Suzuki Y, Yoshimaru T, Inoue T, Takayama T, Ra C (2006) Fungal metabolite gliotoxin blocks mast cell activation by a calcium- and superoxide-dependent mechanism: implications of immunosuppressive activities. Clin Immunol 118:108–116 Nulton CP, Campbell IM (1977) Mycophenolic acid is produced during balanced growth of Penicillium brevicompactum. Can J Microbiol 23:20–27 Nulton CP, Campbell IM (1978) Labelled acetone and levulinic acid are formed when [14C]acetate is being converted to mycophenolic acid in Penicillium brevicompactum. Can J Microbiol 24:1999–2001 Offenzeller M, Su Z, Santer G, Moser H, Traber R, Memmert K, Schneider-Scherzer E (1993) Biosynthesis of the unusual amino acid (4R)-4-[(E)-2-butenyl]4-methyl-L-threonine of cyclosporine A. Identification of 3(R)-hydroxy-4(R)-methyl-6(E)-octenoic acid as a key intermediate by enzymatic in vitro synthesis and by in vivo labelling techniques. J Biol Chem 268: 26127–26134 Ohno N, Miura NN, Chiba N, Adachi Y, Yadomae T (1995) Conparison of the immunopharmacological activities of triple and single-helical schizophyllan in mice. Biol Pharmaceut Bull 18:1242–1247 Oishi T, Ando K, Inomiya K, Sato H, Iida M, Chida N (2002) Total synthesis of (þ)-myriocin and (–)sphingofungin E from aldohexoses using overman rearrangement as the key reaction. Bull Chem Soc Jpn 75:1927–1947 Ooi VEC (2008) Antitumor and immunomodulatory activities of mushroom polysaccharides. In: Cheung PCK (ed) Mushrooms as functional food. Wiley, Singapore, pp 147–198 191 Ooi VEC, Liu F (2000) Immunomodulation and anti-cancer activity of polysaccharide-protein complex. Curr Med Chem 7:715–729 Ozaki H, Ishihara M, Kida T, Yamanaka S, Shibai H (1987a) Mycophenolic acid production by drug-resistant and methionine or glutamic-acid requiring mutants of Penicillium brevicompactum. Agric Biol Chem 51:2509–2514 Ozaki H, Kubota K, Takahashi H (1987b) Effects various adsorbents on mycelium formation and mycophenolic acid production by Penicillium brevicompactum. Agric Biol Chem 51:2503–2508 Pahl HL, Kraub B, Schulze-Osthoff K, Decker T, Traenckner EBM, Vogt M, Myers C, Parks T, Warring P, Mühlbacher A, Czernilofsky A-P, Baeuerle PA (1996) The immunosuppressive fungal metabolite gliotoxin specifically inhibits transcription factor NF-kB. J Exp Med 183:1829–1840 Palleschi A, Bocchinfuso G, Coviello T, Alhaique F (2005) Molecular dynamics investigations of the polysaccharide sclera-glucan: first study on the triple helix structure. Carbohydr Res 340:2154–2162 Patel J, Kobashigawa JA (2008) Minimization of immunosuppression transplant immunology. Transplant Immunol 20:38–54 Patterson JW (1993) The synthesis of mycophenolic acid. Tetrahedron 49:4789–4798 Pirofski L-A, Casadevall A (2006). Immunomodulators as an antimicrobial tool. Curr Opin Microbiol 9: 489–495 Plé PA, Hamon A, Jones, G (1997) A convergent synthesis of mycophenolic acid. Tetrahedron 53:3395–3400 Preddie DC, Markowitz GS, Radhakrishnan J, Nickolas TL, D’Agati VD, Schwimmer JA, Gardenswartz M, Rosen R, Appel GB (2006) Mycophenolate mofetil for the treatment of interstitial nephritis. Clin J Am Soc Nephrol 1:718–722 Puel O, Tadrist S, Galtier P, Oswald I, Delaforge M (2005) Byssochlamys nivea as a source of mycophenolic acid. Appl Environ Microbiol 71:550–553 Queener SW, Nash III CH (1978) Procedure for obtaining Penicillium species mutants with improved ability to synthesize mycophenolic acid. US Patent 4,115,197 Queener SW, Wilkerson SG, Nash III CH (1982) Sterol content and titer of mycophenolic acid in polyene antibiotic resistant mutants of Penicillium stoloniferum. In: Krumphabzl V, Sikyta B, Vanek Z (eds) Overproduction of microbial products. Academic, New York, pp 535–548 Ren Y, Strobel GA, Graff JC, Jutila M, Park SG, Gosh S, Teplow D, Condron M, Pang E, Hess WM, Moore E (2008) Colutellin A, an immunosuppressive peptide from Colletotrichum dematium. Microbiology 154:1973–1979 Research and Markets (2007) http://www.researchandmarkets.com/reports/573667. Accessed 20 May 2009 Reshetnikov SV, Wasser SP, Tan KK (2001) Higher basidiomycetes as a source of antitumor and immunostimulating polysaccharides. Int J Med Mushrooms 3:361–394 192 Hesham El Enshasy Roh YT (2008) Process for mass production of mycophenolic acid by using urea as nitrogen source. Patent WO 2008/026883 A1 Rohloff JC, Gardner JO, Towne RW (1995) Mycophenolate dianions. Tetrahedron Lett 43:7803–7806 Russell R, Paterson M (2008) Cordyceps – a traditional Chinese medicine and another fungal therapeutic biofactory? Phytochemistry 69:1469–1495 Sadhukhan AK, Murthy MVR, Kumar RA, Mohan EVS, Vandana G, Bhar C, Rao KV (1999) Optimization of mycophenolic acid production in solid state fermentation using response surface methodology. J Ind Microbiol 22:33–38 Sakai J-I, Kezuka T, Yokoi H, Okada AA, Usui M, Mizuguchi J, Kaneko Y (1999) Suppressive effects of a novel compound on interphotoreceptor retinoidbinding protein-induced experimental autoimmune uveoretinitis in rats. Allergol Int 48:189–197 Sakamoto K, Tsujii E, Abe F, Nakanishi T, Yamashita M, Shigematsu N, Izumi S, Okuhara M (1996) FR901483, a novel immunosuppressant isolated from Cladobotryum sp. No. 11231. Taxonomy of the producing organism, fermentation, isolation, physico-chemical properties and biological activities. J Antibiot 49:37–44 Sanchis V, Scott PM, Farber JM (1988) Mycotoxin-producing potential of fungi isolated from red kidney beans. Mycopathologia 104:157–162 Sarangi I, Ghosh D, Bhutia SK, Mallick SK, Maiti TK (2006) Anti-tumor and immunomodulating effects of Pleurotus ostreatus mycelia-derived proteoglycans. Int Immunopharmacol 6:1287–1297 Sasaki S, Hashimoto R, Kiuchi M, Inoue K, Ikumoto T, Hirose R, Chiba K, Hoshino Y, Okumoto T, Fujita T (1994) Fungal metabolites. Part 14. Novel potent immunosuppressants, mycestericins, produced by Mycelia sterilia. J Antibiot 47:420–433 Sasaki T, Takasuka N, Chihara G, Maeda YY (1976) Antitumor activity of degraded products of lentinan – its correlation with molecular weight. Gann 67:191–195 Sato H, Sato K, Iida M, Yamanaka H, Oishi T, Chiba N (2008) Total synthesis of mycestericin A. Tetrahedron Lett 49:1943–1947 Sawada H, Kaihara S, Egawa H, Inomata Y, Tanaka K, Yamaoka Y (1996) Efficacy of a new immunosuppressant, CAM, on small bowel transplantation in rats. Transplant Proc 28:2512–2513 Sawai K, Okuno T, Terada Y, Harada Y, Sawamura K, Sasaki H, Takao S (1981) Isolation and properties of two antifungal substances from Fusarium solani. Agric Biol Chem 45:1223–1228 Schneweis I, Meyer K, Hörmansdorfer S, Bauer J (2000) Mycophenolic acid in silage. Appl Environ Microbiol 66:3639–3641 Schreiber SL Crabtree GR (1992) The mechanism of action of cyclosporine A and FK-506. Immunol Today 13:1–15 Seidel G, Laurich D, Fürstner A (2004) Iron-catalyzed cross-coupling reactions. A scalable synthesis of the immunosuppressive agent FTY720. J Org Chem 69:3950–3952 Sekar C, Balaraman K (1998a) Immobilization of the fungus, Tolypocladium sp. for the production of cyclosporine A. Bioproc Eng 18:281–283 Sekar C, Balaraman K (1998b) Optimization studies on the production of cyclosporine A by solid state fermentation. Bioproc Eng 18:293–296 Sekar C, Rajasekar VW, Balaraman K (1997) Production of cyclosporine A by solid state fermentation. Bioproc Eng 17:257–259 Senn H, Weber C, Kobel H, Traber R (1991) Selective 13 C-labelling of cyclosporine A. Eur J Biochem 199:653–658 She QB, Ng TB, Liu WK (1998) A novel lectin with potent immunomodulatory activity isolated from both fruiting bodies and cultured mycelia of the edible mushroom Volvariella volvacea. Biochem Biophys Res Comm 247:106–116 Shibasaki F, Hallin U, Uchino H (2002) Calcineurin as a multifunctional regulator. J Biochem (Tokyo) 131:1–15 Shumpei Y (2002). Mizoribine: mode of action and effects in clinical use. Pediatrics Int 44:196–198 Siekierka JJ, Hung SH, Poe M, Lin CS, Sigal NH (1989) A cytosolic binding protein for the immunosuppressant FK506 has peptidylprolyl isomerase activity but is distinct from cyclophilin. Nature 341: 755–757 Sigg HP, Weber HP (1968) Isolierung und Strukturaufklärung von Ovalicin. Helv Chim Acta 51:1395–1408 Sintchak MD, Fleming MA, Futer O, Raybuck SA, Chambers SP, Caron PR, Murcko MA, Wilson KP (1996) Structure and mechanism of inosine monophosphate dehydrogenase in complex with immunosuppressant mycophenolic acid. Cell 85:921–930 Sircar A, Suryanarayan S, Khedkar AP, Subramaniyam P, Tamber SP (2005) Manufacture and purification of mycophenolic acid. US Patent 6,927,047 Smith JE, Rowan NJ, Sullivan R (2002) Medicinal mushrooms: a rapidly developing area of biotechnology for cancer therapy and other bioactivities. Biotechnol Lett 24:1839–1845 Spatz S, Rundnicka A, McDonald C (1978) Mycophenolic acid in psoriasis. Br J Dermatol 98:429–434 Sugito K, Koshinaga T, Inoue M, Ikeda T, Hagiwara N, Fukuzawa M (2005) The effect of a novel immunosuppressant, FTY720, in mice without secondary lymphoid organs. Surg Today 35:662–667 Survase SA, Saudagar PS, Bajaj IB, Singhal RS (2007) Scleroglucan: fermentative production, downstream processing and applications. Food Technol Biotechnol 45:107–118 Svarstad H, Bugge HC, Dhillion SS (2000) From Norway to Novartis: cyclosporine from Tolypocladium inflatum in an open access bioprospecting regime. Biodiv Conserv 9:1521–1541 Takahashi A, Kusano G, Ohta T, Nozoe S (1988) The constituents of Lactarius flavidulus Imai. Chem Pharm Bull 36:2366–2370 Takahashi A, Kusano G, Ohta T, Nozoe S (1993) Revised structures of flavidulos, constituents of Lactarius flavidulus Imai, and the structure of flavidulol D. Chem Pharm Bull 41:2032–2033 Immunomodulators Takazawa K, Hosoda Y, Bashuda H, Yagita H, Okumura K, Kaneko Y (1995) CAM – a novel immunosuppressive agent. Transplant 59:1723–1727 Takei S (2002) Mizoribine in the treatment of rheumatoid arthritis and juvenile idiopathic arthritis. Pediatr Int 44:205–209 Tanaka E, Inoue E, Kawaguchi Y, Tomatsu T, Yamanaka H, Hara M, Kamatani N (2006) Acceptability and usefuleness of mizoribine in the management of rheumatoid arthritis in methotrexate-refractory patients and elderly patients, based on analysis of data from a large-scale observational cohort study. Mod Rheumatol 16:214–219 Tang YJ, Zhong JJ (2003) Role of oxygen supply in submerged fermentation of Ganoderma lucidum for production of Ganoderma polysaccharide and ganoderic acid. Enz Microb Technol 32:478–484 Tang YJ, Zhu LW, Li HM, Li DS (2007) Submerged culture of mushrooms in bioreactors – challenges, current state-of-the-art, and future prospects. Food Technol Biotechnol 45:221–229 Tao Y, Zhang L, Cheung PCK (2006) Physicochemical properties and antitumor activities of water soluble native and sulfated hyperbranched mushroom polysaccharides. Cabohydr Res 341:2261–2269 Tiwari S, Sathyanathan D, Thomas E, Melarkode R, Ramavana G, Suryanaraya S (2003) Fed batch solid state fermentation for the production of mycophenolic acid. Patent WO/2003/106690 Tönshoff B (2006) Mycophenolsäure nach Nierentransplantation. Nephrologe 1:59–61 Traber R, Loosli HR, Hofmann HR (1982) Isolierung und Strukturermittlung der neuen Cyclosporine E, F, G, H und I. Helv Chem Acta 65:1655–1677 Traber R, Hoffmann H, Lossli HR (1987) Neue Cyclosporine aus Tolypocladium inflatum. Die Cyclosporine K–Z. Helv Chem Acta 70:13–36 Tsukagoshi S, Hashimoto Y, Fujii G, Kobayashi H, Nomoto K, Orita K (1984) Krestin (PSK).Cancer Treat Rev 11:131–155 Turk BE, Su Z, Liu JO (1998) Synthetic analogues of TNP470 and ovalicin reveal a common molecular basis for inhibition of angiogenesis and immunosuppression. Bioorg Med Chem 6:1163–1169 Uchida K, Tominaga Y, Haba T, Katayama A, Matsuoka S, Goto N, Ueki T, Kimata T, Takeda A, Morozumi K, Takagi H, Nakao A (2004) Clinical success of NEORAL absorption profile. Transplant Proc 36 [Suppl 2S]:S461–S464 Van der Hem LG, van der Vliet JA, Kino K, Hoitsma AJ, Tax WJM (1996) Ling Zhi-8: A fungal protein with immunomodulatory effects. Transplant Proc 28:958–959 Velkov T, Lawen A (2003) Non-ribosomal peptide synthetases as technological platforms for the synthesis of highly modified peptide bioeffectors – cyclosporin synthetase as a complex example. Biotechnol Annu Rev 9:151–197 Velkov T, Singaretnam LG, Lawen A (2006) An improved purification procedure for cyclosporine synthetase. Protein Express Purific 45:275–287 193 Vinokurova N, Ivanushkina N, Kochkina G, Arinbasarov M, Ozerskaya S (2005) Production of mycophenolic acid by fungi of genus Penicillium link. Appl Biochem Microbiol 41:83–86 Von Arx JA (1986) Tolypocladium, a synonym of Beauveria. Mycotaxon 25:153–158 Vonderscher J, Meinzer A (1994) Rationale for the development of Sandimmune Neoral. Transplant Proc 26:2925–2927 Wang HX, Liu WK, Ng TB, Ooi VEC, Chang ST (1996) The immunomodulatory and antitumor activities of lectins from mushroom Tricholoma mongolicum. Immunopharmacology 31:205–211 Wang HX, Ng TB, Ooi VEC, Liu WK, Chang ST (1997) Actions of lectins from the mushroom Tricholoma mongolicum on macrophages, splenocytes and lifespan in sarcoma-bearing mice. Anticancer Res 17:419–424 Wang HX, Gao J, Ng TB (2000) A new lectin with highly potent antihepatoma and antisarcoma activities from the oyster mushroom Pleurotus ostreatus. Biochem Biophys Res Comm 275:810–816 Wang JC, Hu SH, Liang ZC, Yeh CJ (2005) Optimization for the production of water-soluble polysaccharide from Pleurotus citrinopileatus in submerged culture and its antitumor effect. Appl Microbiol Biotechnol 67:759–766 Wang P-H, Hsu C-I, Tang SC, Huang Y-L, Lin J-Y, Ko J-L (2004) Fungal immunomodulatory protein from Flammulina velutipes induces interferon-g production through p38 mitogen-activated protein kinase signaling pathway. Agric Food Chem 52:2721–2725 Wang YY, Khoo KH, Chen ST, Lin CC, Wong CH, Lin CH (2002) Studies on the immuno-modulating and antitumor activities of Ganoderma lucidum (Reishi) polysaccharides: functional and proteomic analyses of a fucose-containing glycoprotein fraction responsible for the activites. Bioorg Med Chem 10:1057–1062 Ward C, Chilvers ED, Lawson MF, Pryde JG, Fujihara S, Farrow SN, Haslett C, Rossi AG (1999) NF-kB activation is a critical regulator of human granulocyte apoptosis in vitro. J Biol Chem 274:4309–4318 Waring P, Beaver J (1996) Gliotoxin and related epipolythiodioxopiperazines. Gen Pharmacol 27:1311–1316 Wasser SP (2002) Medicinal mushrooms as a source of antitumor and immunomodulating polysaccharides. Appl Microbiol Biotechnol 60:258–274 Wasser SP, Weis AL (1999) Medicinal properties of substances occurring in higher basidiomycetes mushrooms: current perspectives. Int J Med Mushrooms 1:31–62 Wasser SP, Elisashivili VI, Tan KK (2003) Effects of carbon and nitrogen sources in the medium on Tremella mesenterica Retz.: Fr. (Heterobasidiomycetes) growth and polysaccharide production. Int J Med Mushrooms 5:49–56 Weber G, Leitner E (1994) Disruption of the cyclosporine synthetase gene of Tolypocladium niveum. Curr Gent 26:461–467 Weber G, Schörgendorfer K, Schneider-Scherzer E, Leitner E (1994) The peptide synthetase catalyzing cyclosporine 194 Hesham El Enshasy production in Tolypocladium niveum is encoded by a giant 458-kilobase open reading frame. Curr Genet 26:120–125 Xu ZN, Yang S-T (2007) Production of mycophenolic acid by Penicillium brevicompactum immobilized in a rotating fibrous-bed bioreactor. Enz Microb Technol 40:623–628 Yalin W, Ishurd O, Cuirong S, Yuanjiang P (2005) Structure analysis and anti-tumor activity of (1!3)b-D-glucans (cordyglucans) from the mycelia of Cordyceps sinensis. Planta Medica 71:381–384 Yamada A, Kataoka T, Nagai K (2000) The fungal metabolite gliotoxin: immunosuppressive activity on CTLmediated cytotoxicity. Immunol Lett 71:27–32 Yang B-K, Gu Y-A, Jeong Y-T, Jeong H, Song C-H (2007) Chemical characteristics and immune-modulating activities of exo-biopolymers produced by Grifola frondosa during submerged fermentation process. Int J Biol Macromol 41:327–333 Yang L, Wang R, Liu J, Tong H, Deng Y, Li Q (2004) The effect of polyporus umbellatus polysaccharide on the immunosuppression property of culture supernatant of S180 cells. Chin J Cell Mol Immunol 20:234–237 Yonezawa S, Komurasaki T, Kawada K, Tsuri T, Fuji M, Kugimiya A, Haga N, Mitsumori S, Inagaki M, Nakatani T, Tamura Y, Takechi S, Taishi T, Ohtani M (1998) Total synthesis of terprenin, a novel immunosuppressive p-terphenyl derivative. J Org Chem 63:5831–5837 Zhang M, Cui SW, Cheung PCK, Wang Q (2007) Antitumor polysaccharides from mushrooms: a review on their isolation process, structural characteristics and antitumor activity. Trends Food Sci Technol 18:4–19 Zhang P, Cheung PCK (2002) Evaluation of sulfated Lentinus edodes a-(1!3)-D-glucan as a potential antitumor agent. Biosci Biotechnol Biochem 66: 1052–1056 Zhang WY, Wang Y, Hou YY (2004) Effects of Chinese medicinal fungus water extract on tumor metastasis and some parameters of immune function. Int Immunopharmacol 4:461–468 Zhong JJ, Tang YJ (2004) Submerged cultivation of medicinal mushrooms for production of valuable bioactive metabolites. Adv Biochem Eng Biotechnol 87:25–59 Zou X (2006) Fed-batch fermentation for hyperproduction of polysaccharide and ergosterol by medicinal mushroom Agaricus brasiliensis. Process Biochem 41: 970–974