Skip to main content

Part of the book series: Springer Handbooks ((SHB))

Abstract

Marine-derived metabolites continue to be a prolific source for bioactive natural products with a high tendency to become drug candidates. Recently, eribulin mesylate (GlossaryTerm

E7389

), a synthetic derivative of halichondrin B, was approved by the US GlossaryTerm

FDA

(Food and Drug Administration) for the treatment of breast cancer metastases, going under the trade name Halaven. Some other marine-derived pharmaceuticals have likewise already been approved and launched onto the market, including Yondelis (trabectedin), Prialt (ziconotide), Retrovir (zidovudine), Cytosar (cytarabine, GlossaryTerm

Ara-C

), and Vira-A (vidarabine, Ara-A). Research interests in marine natural products have increased in the last few decades, in particular with respect to compounds isolated from marine-derived fungi. In this chapter, a historical background of marine-derived pharmaceuticals and fungal metabolites will be presented, including potential classes of bioactive chemical compounds. In addition, a perspective on the sustainability of marine-derived fungi with regard to a continuous supply of bioactive compounds for market needs rather than a pursuit of other strategies such as aquafarming will be discussed.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 269.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 349.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Abbreviations

1-D:

one-dimensional

2-D:

two-dimensional

A549:

human lung adenocarcinoma

AChE:

acetylcholinesterase

AIDS:

acquired immunodeficiency syndrome

ALCL:

anaplastic large cell lymphoma

ATCC:

American Type Culture Collection

Ara-A:

arabinofuranosyl adenine or adenine arabinoside

Ara-C:

arebinosyl cytosine

cytarabine

CD:

circular dichroism

CHO:

Chinese hamster ovary

CML:

chronic myeloid leukemia

CNS:

central nervous system (CNS)

Campto:

camptothecin

DMXBA:

3-(2,4-dimethoxybenzylidene)-anabaseine

DNA:

deoxyribonucleic acid

DPPH:

1,1-diphenyl-2-picrylhydrazyl

E7389:

eribulin mesylate

ECD:

electronic circular dichroism

EMEA:

European Medicines Agency

ET-743:

ecteinascidins

EtOAc:

ethyl acetate

FDA:

Food and Drug Administration

HBV:

hepatitis B virus

HCT:

human colorectal tumor

HDAC:

histone deacetylase

HIV:

human immunodeficiency virus

HLE:

human leukocyte elastase

HMT:

histone methyltransferases

HPLC:

high-performance liquid chromatography

HUVEC:

human umbilical vein endothelial cells

HeLa:

human cervix carcinoma cells

Hep3B:

human liver carcinoma

HepG2:

Hepatoma Growth2

IMPDH:

inosine 5 -monophosphate dehydrogenase

K562:

human chronic myeloid leukemia cells

KB:

human epidermoid carcinoma

LPS:

lipopolysaccharide

MA:

maslinic acid

MCF:

Michigan Cancer Foundation-7

MIC:

minimum inhibitory concentration

MMAE:

monomethyl auristatin E

MOLT:

molybdate uptake transporter

MPA:

mycophenolic acid

MRSA:

methicillin-resistant Staphylococcus aureus

MS:

mass spectroscopy

MTT:

microculture tetrazolium assay

M:

mitosis

NCI-H460:

human lung cancer cell line

NMR:

nuclear magnetic resonance

NOESY:

nuclear Overhauser and exchange spectroscopy

NRPS:

nonribosomal peptide synthase

NSCLC:

non-small cell lung cancer

PC:

phycocyanin

PKS:

polyketide synthase

PRRSV:

porcine reproductive and respiratory syndrome virus

PTP1B:

protein tyrosine phosphatase 1B

RAW:

ATCC cell line

RT:

reverse transcriptase

SAHA:

suberoylanilide hydroxamic acid

SAR:

structure-activity relationship

SF-268:

human CNS cancer cell line

SF:

surfactin

SIV:

subintestinal vessel plexus

S:

synthesis

TDDFT:

time-dependent density functional theory

VDA:

vascular disrupting agent

iNOS:

inducible nitric oxide synthase

References

  1. A.L. Harvey: Natural products in drug discovery, Drug Discov. Today 13, 894–901 (2008)

    Article  CAS  Google Scholar 

  2. T.F. Molinski, D.S. Dalisay, S.L. Lievens, J.P. Saludes: Drug development from marine natural products, Nat. Rev. Drug Discov. 8, 69–85 (2009)

    Article  CAS  Google Scholar 

  3. A.M. Mayer, K.B. Glaser, C. Cuevas, R.S. Jacobs, W. Kem, R.D. Little, J.M. McIntosh, D.J. Newman, B.C. Potts, D.E. Shuster: The odyssey of marine pharmaceuticals: A current pipeline perspective, Trends Pharmacol. Sci. 31, 255–265 (2010)

    Article  CAS  Google Scholar 

  4. S.S. Ebada, P. Proksch: Marine organisms and their prospective use in therapy of human diseases. In: Nature Helps $\ldots$ . How Plants and Other Organisms Contribute to Solve Health Problems, ed. by H. Melhorn (Springer, Berlin, Heidelberg 2011) pp. 153–190

    Google Scholar 

  5. A.R. Duckworth, G.A. Samples, A.E. Wright, S.A. Pomponi: In vitro culture of the ascidian Ecteinascidia turbinata to supply the antitumor compounds ecteinascidins, Aquaculture 241, 427–439 (2004)

    Article  CAS  Google Scholar 

  6. W. Bergmann, R.J. Feeney: Contributions to the study of marine products. XXXII, The nucleosides of sponges. I, J. Org. Chem. 16, 981–987 (1951)

    Article  CAS  Google Scholar 

  7. W. Bergmann, R.J. Feeney: The isolation of a new thymine pentoside from sponges, J. Am. Chem. Soc. 72, 2809–2810 (1950)

    Article  CAS  Google Scholar 

  8. L. Zhang, R. An, J. Wang, N. Sun, S. Zhang, J. Hu, J. Kuai: Exploring novel bioactive compounds from marine microbes, Curr. Opin. Microbiol. 8, 276–281 (2005)

    Article  CAS  Google Scholar 

  9. T. Mavromoustakos, T. Calogeropoulou, M. Koufaki, A. Kolocouris, I. Daliani, C. Demetzos, Z. Meng, A. Makriyannis, J. Balzarini, E. De Clercq: Ether phospholipid-AZT conjugates possessing anti-HIV and antitumor cell activity. Synthesis, conformational analysis, and study of their thermal effects on membrane bilayers, J. Med. Chem. 44, 1702–1709 (2001)

    Article  CAS  Google Scholar 

  10. R. Yarchoan, R. Klecker, K. Weinhold, P.D. Markham, H.K. Lyerly, D.T. Durack, E. Gelmann, S.N. Lehrman, R.M. Blum, D.W. Barry: Administration of 3'-azido-3'-deoxythymidine, an inhibitor of HTLV-III/LAV replication, to patients with AIDS or AIDS-related complex, Lancet 1, 575–580 (1986)

    Article  CAS  Google Scholar 

  11. H. Mitsuya, R. Yarchoan, S. Broder: Molecular targets for AIDS therapy, Science 249, 1533–1544 (1990)

    Article  CAS  Google Scholar 

  12. J. Balzarini, G.J. Kang, M. Dalal, P. Herdewijn, E. De Clercq, S. Broder, D.G. Johns: The anti-HTLV-III (anti-HIV) and cytotoxic activity of 2',3'-didehydro-2',3'-dideoxyribonucleosides: A comparison with their parental 2',3'-dideoxyribonucleosides, Mol. Pharmacol. 32, 162–167 (1987)

    CAS  Google Scholar 

  13. R.F. Schinazi, C.K. Chu, A. Peck, A. McMillan, R. Mathis, D. Cannon, L.S. Jeonq, J.W. Beach, W.B. Choi, S. Yeola: Activities of the four optical isomers of 2',3'-dideoxy-3'-thiacytidine (BCH-189) against human immunodeficiency virus type 1 in human lymphocytes, Antimicrob. Agents Chemother. 36, 672–676 (1992)

    Article  CAS  Google Scholar 

  14. B.M. Olivera, W.R. Gray, R. Zeikus, J.M. McIntosh, J. Varga, J. Rivier, V. de Santos, L.J. Cruz: Peptide neurotoxins from fish-hunting cone snails, Science 230, 1338–1343 (1985)

    Article  CAS  Google Scholar 

  15. H. Terlau, B.M. Olivera: Conus venoms: A rich source of novel ion channel-targeted peptides, Physiol. Rev. 84, 41–68 (2004)

    Article  CAS  Google Scholar 

  16. S.S. Ebada, R.A. Edrada, W. Lin, P. Proksch: Methods for isolation, purification, and structural elucidation of bioactive secondary metabolites from marine invertebrates, Nature Protoc. 3, 1820–1831 (2008)

    Article  CAS  Google Scholar 

  17. D. Mendola: Aquaculture production of bryostatin 1 and ecteinascidin 743. In: Drugs from the Sea, ed. by N. Fusetani (Karger, Basel 2000) pp. 120–133

    Chapter  Google Scholar 

  18. D. Mendola: Aquaculture of three phyla of marine invertebrates to yield bioactive metabolites: Process developments and economies, Biomol. Eng. 20, 441–458 (2003)

    Article  CAS  Google Scholar 

  19. C. Cuevas, M. Pérez, M.J. Martín, J.L. Chicharro, C. Fernández-Rivas, M. Flores, A. Francesch, P. Gallego, M. Zarzuelo, F. de La Calle, J. García, C. Polanco, I. Rodríguez, I. Manzanares: Synthesis of ecteinascidin ET-743 and phthalascidin Pt-650 from cyanosafracin B, Org. Lett. 2, 2545–2548 (2000)

    Article  CAS  Google Scholar 

  20. D. Uemura, K. Takahashi, T. Yamamoto: Norhalichondrin A: An antitumor polyether macrolide from a marine sponge, J. Am. Chem. Soc. 107, 4796–4798 (1985)

    Article  CAS  Google Scholar 

  21. Y. Hirata, D. Uemura: Halichondrins–antitumor polyether macrolides from a marine sponge, Pure Appl. Chem. 58, 701–710 (1986)

    Article  CAS  Google Scholar 

  22. G.R. Pettit: The dolastatins, Prog. Chem. Org. Nat. Prod. 70, 1–79 (1997)

    CAS  Google Scholar 

  23. R. Bai, G.R. Pettit, E. Hamel: Dolastatin 10, a powerful cytostatic peptide derived from a marine animal – Inhibition of tubulin polymerization mediated through the vinca alkaloid binding domain, Biochem. Pharmacol. 39, 1941–1949 (1990)

    Article  CAS  Google Scholar 

  24. R. Bai, G.R. Pettit, E. Hamel: Binding of dolastatin 10 to tubulin at a distinct site for peptide antimitotic agents near the exchangeable nucleotide and vinca alkaloid sites, J. Biol. Chem. 265, 17141–17149 (1990)

    CAS  Google Scholar 

  25. S.O. Doronina, B.E. Toki, M.Y. Torgov, B.A. Mendelsohn, C.G. Cerveny, D.F. Chace, R.L. DeBlanc, R.P. Gearing, T.D. Bovee, C.B. Siegall, J.A. Francisco, A.F. Wahl, D.L. Meyer, P.D. Senter: Development of potent monoclonal antibody auristatin conjugates for cancer therapy, Nature Biotechnol. 21, 778–784 (2003)

    Article  CAS  Google Scholar 

  26. J.A. Francisco, C.G. Cerveny, D.L. Meyer, B.J. Mixan, K. Klussman, D.F. Chace, S.X. Rejniak, K.A. Gordon, R. DeBlanc, B.E. Toki, C.-L. Law, S.O. Doronina, C.B. Siegall, P.D. Senter, A.F. Wahl: cAC10-vcMMAE, an anti-CD30-monomethyl auristatin E conjugate with potent and selective antitumor activity, Blood 102, 1458–1465 (2003)

    Article  CAS  Google Scholar 

  27. A. Younes, U. Yasothan, P. Kirkpatrick: Brentuximab vedotin, Nat. Rev. Drug Discov. 11, 19–20 (2012)

    Article  CAS  Google Scholar 

  28. K.L. Rinehart, A.M. Lithgow-Bertelloni: Dehydrodidemnin B, WO9104985 (1991)

    Google Scholar 

  29. K.L. Rinehart, J.B. Gloer, J.C. Cook, S.A. Mizsak, T.A. Scahill: Structures of the didemnins, antiviral and cytotoxic depsipeptides from a Caribbean tunicate, J. Am. Chem. Soc. 103, 1857–1859 (1981)

    Article  CAS  Google Scholar 

  30. O. Kucuk, M.L. Young, T.M. Habermann, B.C. Wolf, J. Jimeno, P.A. Cassileth: Phase II trial of didemnin B in previously treated non-Hodgkin's lymphoma: An Eastern cooperative oncology group (ECOG) study, Am. J. Clin. Oncol. 23, 273–277 (2000)

    Article  CAS  Google Scholar 

  31. B. Nuijen, M. Bouma, C. Manada, J.M. Jimeno, J.H.M. Schellens, A. Bult, J.H. Beijnen: Pharmaceutical development of anticancer drugs derived from marine sources, Anticancer Drugs 11, 793–811 (2000)

    Article  CAS  Google Scholar 

  32. C.S. Mitsiades, E.M. Ocio, A. Pandiella, P. Maiso, C. Gajate, M. Garayoa, D. Vilanova, J.C. Montero, N. Mitsiades, C.J. McMullan, N.C. Munshi, T. Hideshima, D. Chauhan, P. Aviles, G. Otero, G. Faircloth, M.V. Mateos, P.G. Richardson, F. Mollinedo, J.F. San-Miguel, K.C. Anderson: Aplidin, a marine organism-derived compound with potent antimyeloma activity in vitro and in vivo, Cancer Res. 68, 5216–5225 (2008)

    Article  CAS  Google Scholar 

  33. C. Le Tourneau, E. Raymond, S. Faivre: Aplidine: A paradigm of how to handle the activity and toxicity of a novel marine anticancer poison, Curr. Pharm. Des. 13, 3427–3439 (2007)

    Article  Google Scholar 

  34. W. Kem, F. Soti, K. Wildeboer, S. LeFrancois, L. MacDougall, D.-Q. Wei, K.-C. Chou, H.R. Arias: The nemertine toxin anabaseine and its deriative DMXBA (GTS-21): Chemical and pharmacological properties, Mar. Drugs 4, 255–273 (2006)

    Article  CAS  Google Scholar 

  35. J.J. Buccafusco, S.R. Letchworth, M. Bencherif, P.M. Lippiello: Long-lasting cognitive improvement with nicotinic receptor agonists: mechanisms of pharmacokinetic-pharmacodynamic discordance, Trends Pharmacol. Sci. 26, 352–360 (2005)

    Article  CAS  Google Scholar 

  36. H. Kitagawa, T. Takenouchi, R. Azuma, K.A. Wesnes, W.G. Kramer, D.E. Clody, A.L. Burnett: Safety, pharmacokinetics, and effects on cognitive function of multiple doses of GTS-21 in healthy, male volunteers, Neuropsychopharmacology 28, 542–551 (2003)

    Article  CAS  Google Scholar 

  37. A. Olincy, J.G. Harris, L.L. Johnson, V. Pender, S. Kongs, D. Allensworth, K. Ellis, G.O. Zerbe, S. Leonard, K.E. Stevens, J.O. Stevens, L. Martin, L.E. Adler, F. Soti, W.R. Kem, R. Freedman: Proof-of-concept trial of an α7 nicotinic agonist in schizophrenia, Arch. Gen. Psychiatr. 63, 630–638 (2006)

    Article  CAS  Google Scholar 

  38. R. Freedman, A. Olincy, R.W. Buchanan, J.G. Harris, J.M. Gold, L. Johnson, D. Allensworth, A. Guzman-Bonilla, B. Clement, M.P. Ball, J. Kutnick, V. Pender, L.F. Martin, K.E. Stevens, B.D. Wagner, G.O. Zerbe, F. Soti, W.R. Kem: Initial Phase 2 trial of a nicotinic agonist in schizophrenia, Am. J. Psych. 165, 1040–1047 (2008)

    Article  Google Scholar 

  39. B. Nicholson, G.K. Lloyd, B.R. Miller, M.A. Palladino, Y. Kiso, Y. Hayashi, S.T. Neuteboom: NPI-2358 is a tubulin-depolymerizing agent: in-vitro evidence for activity as a tumor vascular-disrupting agent, Anticancer Drugs 17, 25–31 (2006)

    Article  CAS  Google Scholar 

  40. Y. Yamazaki, K. Kohno, H. Yasui, Y. Kiso, M. Akamatsu, B. Nicholson, G. Deyanat-Yazdi, S. Neuteboom, B. Potts, G.K. Lloyd, Y. Hayashi: Tubulin photoaffinity labeling with biotin-tagged derivatives of potent diketopiperazine antimicrotubule agents, ChemBioChem 9, 3074–3081 (2008)

    Article  CAS  Google Scholar 

  41. P.J. Scheuer, M.T. Hamann: Kahalalide F: A bioactive depsipeptide from the sacoglossan mollusk Elysia rufescens and the green alga Bryopsis sp., J. Am. Chem. Soc. 115, 5825–5826 (1993)

    Article  Google Scholar 

  42. Y.H. Ling, M. Aracil, J. Jimeno, R. Perez-Soler, Y. Zou: Molecular pharmacodynamics of PM02734 (elisidepsin) as single agent and in combination with erlotinib; synergisitic activity in human non-small cell lung cancer cell lines and xenograft models, Eur. J. Cancer 45, 1855–1864 (2009)

    Article  CAS  Google Scholar 

  43. J.M. Frincke, D.J. Faulkner: Antimicrobial metabolites of the sponge Reniera sp., J. Am. Chem. Soc. 104, 265–269 (1982)

    Article  CAS  Google Scholar 

  44. H.Y. He, D.J. Faulkner: Renieramycins E and F from the sponge Reniera sp.: Reassignment of stereochemistry of the renieramycins, J. Org. Chem. 54, 5822–5824 (1989)

    Article  CAS  Google Scholar 

  45. A. Fontana, P. Cavaliere, S. Wahidulla, C.G. Naik, G. Cimino: A new antitumor isoquinoline alkaloid from the marine nudibranch Jorunna funebris, Tetrahedron 56, 7305–7308 (2000)

    Article  CAS  Google Scholar 

  46. J.F.M. Leal, V. García-Hernández, V. Moneo, A. Domingo, J.A. Bueren-Calabuig, A. Negri, F. Gago, M.J. Guillén-Navarro, P. Avilés, C. Cuevas, L.F. García-Fernández, C.M. Galmarini: Molecular pharmacology and antitumor activity of Zalypsis in several human cancer cell lines, Biochem. Pharmacol. 78, 162–170 (2009)

    Article  CAS  Google Scholar 

  47. R. Bai, S.J. Friedman, G.R. Pettit, E. Hamel: Dolastatin 15, a potent antimitotic depsipeptide derived from Dolabella auricularia – Interaction with tubulin and effects on cellular microtubules, Biochem. Pharmacol. 43, 2637–2645 (1992)

    Article  CAS  Google Scholar 

  48. A. Ray, T. Okouneva, T. Manna, H.P. Miller, S. Schmid, L. Arthaud, R. Luduena, M.A. Jordan, L. Wilson: Mechanism of action of the microtubule-targeted antimitotic depsipeptide tasidotin (formerly ILX651) and its major metabolite tasidotin C-carboxylate, Cancer Res. 67, 3767–3776 (2007)

    Article  CAS  Google Scholar 

  49. R. Bai, M.C. Edler, P.L. Bonate, T.D. Copeland, G.R. Pettit, R.F. Luduena, E. Hamel: Intracellular activation and deactivation of tasidotin, an analog of dolastatin 15: Correlation with cytotoxicity, Mol. Pharmacol. 75, 218–226 (2009)

    Article  CAS  Google Scholar 

  50. R.H. Feling, G.O. Buchanan, T.J. Mincer, C.A. Kauffman, P.R. Jensen, W. Fenical: Salinosporamide A: A highly cytotoxic proteasome inhibitor from a novel microbial source, a marine bacterium of the new genus Salinospora, Prog. Chem. Org. Nat. Prod. 42, 355–357 (2003)

    CAS  Google Scholar 

  51. W. Fenical, P.R. Jensen, M.A. Palladino, K.S. Lam, G.K. Lloyd, B.C. Potts: Discovery and development of the anticancer agent salinosporamide A (NPI-0052), Bioorg. Med. Chem. 17, 2175–2180 (2009)

    Article  CAS  Google Scholar 

  52. M. Groll, R. Huber, B.C.M. Potts: Crystal structures of salinosporamide A (NPI-0052) and B (NPI-0047) in complex with the 20S proteasome reveal important consequences of β-lactone ring opening and a mechanism for irreversible binding, J. Am. Chem. Soc. 128, 5136–5141 (2006)

    Article  CAS  Google Scholar 

  53. D. Chauhan, L. Catley, G. Li, K. Podar, T. Hideshima, M. Velankar, C. Mitsiades, N. Mitsiades, H. Yasui, A. Letai, H. Ovaa, C. Berkers, B. Nicholson, T.H. Chao, S.T. Neuteboom, P. Richardson, M.A. Palladino, K.C. Anderson: A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from bortezomib, Cancer Cell 8, 407–419 (2005)

    Article  CAS  Google Scholar 

  54. D. Chauhan, A. Singh, M. Brahmandam, K. Podar, T. Hideshima, P. Richardson, N. Munshi, M.A. Palladino, K.C. Anderson: Combination of proteasome inhibitors bortezomib and NPI-0052 trigger in vivo synergistic cytotoxicity in multiple myeloma, Blood 111, 1654–1664 (2008)

    Article  CAS  Google Scholar 

  55. R. Talpir, Y. Benayahu, Y. Kashman, L. Pannell, M. Schleyer: Hemiasterlin and geodiamolide TA; two new cytotoxic peptides from the marine sponge Hemiasterella minor (Kirkpatrick), Tetrahedron Lett. 35, 4453–4456 (1994)

    Article  CAS  Google Scholar 

  56. J.E. Coleman, E.D. de Silva, F. Kong, R.J. Andersen: Cytotoxic peptides from the marine sponge Cymbastela sp., Tetrahedron 51, 10653–10662 (1995)

    Article  CAS  Google Scholar 

  57. W.R. Gamble, N.A. Durso, R.W. Fuller, C.K. Westergaard, T.R. Johnson, D.L. Sackett, E. Hamel, J.H. Cardellina, M.R. Boyd: Cytotoxic and tubulin-interactive hemiasterlins from Auletta sp. and Siphonochalina sp. sponges, Bioorg. Med. Chem. 7, 1611–1615 (1999)

    Article  CAS  Google Scholar 

  58. H.J. Anderson, J.E. Coleman, R.J. Andersen, M. Roberge: Cytotoxic peptides hemiasterlin, hemiasterlin A and hemiasterlin B induce mitotic arrest and abnormal spindle formation, Cancer Chemother. Pharmacol. 39, 223–226 (1997)

    Article  CAS  Google Scholar 

  59. J.E. Coleman, B.O. Patrick, R.J. Andersen, S.J. Rettig: Hemiasterlin methyl ester, Acta Crystallogr. C52, 1525–1527 (1996)

    CAS  Google Scholar 

  60. F. Loganzo, C.M. Discafani, T. Annable, C. Beyer, S. Musto, M. Hari, X. Tan, C. Hardy, R. Hernandez, M. Baxter, C. Singanallore, G. Khafizova, M.S. Poruchynsky, T. Fojo, J.A. Nieman, S. Ayral-Kaloustain, A. Zask, R.J. Andersen, L.M. Greenberger: Antimirotubule agent that circumvents P-glycoprotein-mediated resistance in vitro and in vivo, Cancer Res. 63, 1838–1845 (2003)

    CAS  Google Scholar 

  61. M.J. Ratain, S. Undevia, L. Janisch, S. Roman, P. Mayer, M. Buckwalter, D. Foss, B.L. Hamilton, J. Fischer, R.M. Bukowski: Phase 1 and pharmacological study of HTI-286, a novel antimicrotubule agent: Correlation of neutropenia with time above a threshold serum concentration, Proc. Am. Soc. Clin. Oncol. 22, 516 (2003)

    Google Scholar 

  62. B.A. Hadaschik, S. Ettinger, R.D. Sowery, A. Zoubeidi, R.J. Andersen, M. Roberge, M.E. Gleave: Targeting prostate cancer with HTI-286, a synthetic analogue of the marine sponge product hemiasterlin, Int. J. Cancer 122, 2368–2376 (2008)

    Article  CAS  Google Scholar 

  63. G.R. Pettit, C.L. Herald, D.L. Doubek, D.L. Herald, E. Arnold, J. Clardy: Isolation and structure of bryostatin 1, J. Am. Chem. Soc. 104, 6846–6848 (1982)

    Article  CAS  Google Scholar 

  64. G.R. Pettit: Progress in the discovery of biosynthetic anticancer drugs, J. Nat. Prod. 59, 812–821 (1996)

    Article  CAS  Google Scholar 

  65. M.A. Rouhi: Supply issues complicate track of chemicals from sea to market, Chem. Eng. News 73, 42–44 (1995)

    Article  Google Scholar 

  66. L.H. Cragg, M. Andreeff, E. Feldman, J. Roberts, A. Murgo, M. Winning, M.B. Tombes, G. Roboz, L. Kramer, S. Grant: Phase I trial and correlative laboratory studies of bryostatin 1 (NSC 339555) and high-dose 1-β-D-arabinofuranosylcytosine in patients with refractory acute leukemia, Clin. Cancer Res. 8, 2123–2133 (2002)

    CAS  Google Scholar 

  67. A. Dowlati, H.M. Lazarus, P. Hartman, J.W. Jacobberger, C. Whitacre, S.L. Gerson, P. Ksenich, B.W. Cooper, P.S. Frisa, M. Gottlieb, A.J. Murgo, S.C. Remick: Phase I and correlative study of combination bryostatin 1 and vincristine in relapsed B-cell malignancies, Clin. Cancer Res. 9, 5929–5935 (2003)

    CAS  Google Scholar 

  68. J.A. Ajani, Y. Jiang, J. Faust, B.B. Chang, L. Ho, J.C. Yao, S. Rousey, S. Dakhil, R.C. Cherny, C. Craig, A. Bleyer: A multi-center phase II study of sequential paclitaxel and bryostatin 1 (NSC 339555) in patients with untreated, advanced gastric or gastroesophageal junction adenocarcinoma, Investig. New Drugs 24, 353–357 (2006)

    Article  CAS  Google Scholar 

  69. A.M.S. Mayer, K.R. Gustafson: Marine pharmacology in 2005–2006: Antitumor and cytotoxic compounds, Eur. J. Cancer 44, 2357–2387 (2008)

    Article  CAS  Google Scholar 

  70. A.M.S. Mayer, A.D. Rodríguez, R.G.S. Berlinck, M.T. Hamann: Marine pharmacology in 2005–6: Marine compounds with anthelmintic, antibacterial, anticoagulant, antifungal, anti-inflammatory, antimalarial, antiprotozoal, antituberculosis, and antiviral activities; affecting the cardiovascular, immune and nervous systems, and other miscellaneous mechanisms of action, Biochim. Biophys. Acta 1790, 283–402 (2009)

    Article  CAS  Google Scholar 

  71. M.S. Laport, O.C.S. Santos, G. Muricy: Marine sponges: Potential sources of new antimicrobial drugs, Curr. Pharm. Biotechnol. 10, 86–105 (2009)

    Article  CAS  Google Scholar 

  72. A. Staniek, H.J. Woerdenbag, O. Kayser: Taxomyces andreanae: A presumed paclitaxel producer demystified?, Planta Medica 75, 1561–1566 (2009)

    Article  CAS  Google Scholar 

  73. T. Amna, S.C. Purl, V. Verma, J.P. Sharma, R.K. Khajuria, J. Musarrat, M. Spiteller, G.N. Qazi: Bioreactor studies on the endophytic fungus Entrophospora infrequens for the production of an anticancer alkaloid camptothecin, Can. J. Microbiol. 52, 189–196 (2006)

    Article  CAS  Google Scholar 

  74. V. Rukachaisirikul, N. Khamthong, Y. Sukpondma, C. Pakawatchai, S. Phongpaichit, J. Sakayaroj, K. Kirtikara: An [11]cytochalasin derivative from the marine-derived fungus Xylaria sp. PSU-F100, Chem. Pharm. Bull. 57, 1409–1411 (2009)

    Article  CAS  Google Scholar 

  75. Z. Chen, H. Huang, Y. Chen, Z. Wang, J. Ma, B. Wang, W. Zhang, C. Zhang, J. Ju: New cytochalasins from the marine-derived fungus Xylaria sp. SCSIO 156, Helv. Chim. Acta 94, 1671–1676 (2011)

    Article  CAS  Google Scholar 

  76. S.A. Neff, S.U. Lee, Y. Asami, J.S. Ahn, H. Oh, J. Baltrusaitis, J.B. VGloer, D.T. Wicklow: Aflaquinolones A–G: secondary metabolites from marine and fungicolous isolates of Aspergillus spp., J. Nat. Prod. 75, 464–472 (2012)

    Article  CAS  Google Scholar 

  77. D. Zhang, M. Satake, S. Fukuzawa, K. Sugahara, A. Niitsu, T. Shirai, K. Tachibana: Two new indole alkaloids, 2-(3,3-dimethylprop-1-ene)-costaclavine and 2-(3,3-dimethylprop-1-ene)-epicostaclavine, from the marine-derived fungus Aspergillus fumigatus, J. Nat. Med. 66, 222–226 (2012)

    Article  CAS  Google Scholar 

  78. L. Ding, H.-M. Dahse, C. Hertweck: Cytotoxic alkaloids from Fusarium incarnatum associated with the mangrove tree Aegiceras coniculatum, J. Nat. Prod. 75, 617–621 (2012)

    Article  CAS  Google Scholar 

  79. C. Almeida, N. Part, S. Bouhired, S. Kehraus, G.M. König: Stachylines A–D from the sponge-derived fungus Stachylidium sp., J. Nat. Prod. 74, 21–25 (2011)

    Article  CAS  Google Scholar 

  80. C.-S. Li, C.-Y. An, X.-M. Li, S.-S. Gao, C.-M. Cui, H.-F. Sun, B.-G. Wang: Triazole and dihydroimidazole alkaloids from the marine sediment-derived fungus Penicillium paneum SD-44, J. Nat. Prod. 74, 1331–1334 (2011)

    Article  CAS  Google Scholar 

  81. F. Zhu, G. Chen, X. Chen, M. Huang, X. Wan: Aspergicin, a new antibacterial alkaloid produced by mixed fermentation of two marine-derived mangrove epiphytic fungi, Chem. Nat. Comp. 47, 767–769 (2011)

    Article  CAS  Google Scholar 

  82. E. Julianti, H. Oh, H.-S. Lee, D.-C. Oh, K.-B. Oh, J. Shin: Acremolin, a new 1H-azirine metabolite from the marine-derived fungus Acremonium strictum, Tetrahedron Lett. 53, 2885–2886 (2012)

    Article  CAS  Google Scholar 

  83. C. Almeida, Y. Hemberger, S.M. Schmitt, S. Bouhired, L. Natesan, S. Kehraus, K. Dimas, M. Gütschow, G. Bringmann, G.M. König: Marilines A–C: novel phthalimidines from the sponge-derived fungus Stachylidium sp., Chem. Eur. J. 18, 8827–8834 (2012)

    Article  CAS  Google Scholar 

  84. W.C. Groustas, D. Dou, K.R. Alliston: Neutrophil elastase inhibitors, Expert Opin. Therap. Patents 21, 339–354 (2011)

    Article  CAS  Google Scholar 

  85. B. Korkmaz, M.S. Horwitz, D.E. Jenne, F. Gauthier: Neutrophil elastase, Proteinase 3, and cathepsin G as therapeutic targets in human diseases, Pharmacol. Rev. 62, 726–759 (2010)

    Article  CAS  Google Scholar 

  86. M.F. Elsebai, L. Natesan, S. Kehraus, I.E. Mohamed, G. Schnakenburg, F. Sasse, S. Shaaban, M. Gütschow, G.M. König: HLE-inhibitory alkaloids with a polyketide skeleton from the marine-derived fungus Coniothyrium cereal, J. Nat. Prod. 74, 2282–2285 (2011)

    Article  CAS  Google Scholar 

  87. F. Song, B. Ren, K. Yu, C. Chen, H. Guo, N. Yang, H. Gao, X. Liu, M. Liu, Y. Tong, H. Dai, H. Bai, J. Wang, L. Zhang: Quinazolin-4-one coupled with pyrrolidin-2-iminium alkaloids from marine-derived fungus Penicillium aurantiogriseum, Mar. Drugs 10, 1297–1306 (2012)

    Article  CAS  Google Scholar 

  88. L. Chen, K. Huang, P. Zhong, X. Hu, Z.-X. Fang, J.-L. Wu, Q.-Q. Zhang: Tumonoic acids K and L, novel metabolites from the marine-derived fungus Penicillium citrinum, Heterocycles 85, 413–419 (2012)

    Article  CAS  Google Scholar 

  89. S.S. Afiyatullov, O.I. Zhuravleva, A.S. Antonov, A.I. Kalinovsky, M.V. Pivkin, E.S. Menchinskaya, D.L. Aminin: New metabolites from the marine-derived fungus Aspergillus fumigatus, Nat. Prod. Commun. 7, 497–500 (2012)

    CAS  Google Scholar 

  90. M. El Amrani, A. Debbab, A.H. Aly, V. Wray, S. Dobrestov, W.E.G. Müller, W. Lin, D. Lai, P. Proksch: Farinomalein derivatives from an unidentified endophytic fungus isolated from the mangrove plant Avicennia marina, Tetrahedron Lett. 53, 6721–6724 (2012)

    Article  CAS  Google Scholar 

  91. H.-B. Liu, R.A. Edrada-Ebel, R. Ebel, Y. Wang, B. Schulz, S. Draeger, W.E.G. Müller, V. Wray, W.-H. Lin, P. Proksch: Ophiobolin sesterterpenoids and pyrrolidine alkaloids from the sponge-derived fungus Aspergillus ustus, Helv. Chim. Acta 94, 623–631 (2011)

    Article  CAS  Google Scholar 

  92. M.F. Elsebai, V. Rempel, G. Schnakenburg, S. Kehraus, C.E. Müller, G.M. König: Identification of a potent and selective cannabinoid CB${}_{{1}}$ receptor antagonist from Auxarthron reticulatum, ACS Med. Chem. Lett. 2, 866–869 (2011)

    Article  CAS  Google Scholar 

  93. F.-Y. Du , X.-M. Li, C.-S. Li, Z. Shang, B.-G. Wang: Cristatumins A–D, new indole alkaloids from the marine-derived endophytic fungus Eurotium cristatum EN-220, Bioorg. Med. Chem. Lett. 22, 4650–4652 (2012)

    Article  CAS  Google Scholar 

  94. H.-J. Yan, X.-M. Li, C.-S. Li, B.-G. Wang: Alkaloid and anthraquinone derivatives produced by the marine-derived endophytic fungus Eurotium rubrum, Helv. Chim. Acta 95, 163–168 (2012)

    Article  CAS  Google Scholar 

  95. F. He, Y.-L. Sun, K.-S. Liu, X.-Y. Zhang, P.-Y. Qian, Y.-F. Wang, S.-H. Qi: Indole alkaloids from marine-derived fungus Aspergillus sydowii SCSIO 00305, J. Antibiot. 65, 109–111 (2012)

    Article  CAS  Google Scholar 

  96. S.-U. Lee, Y. Asami, D. Lee, J.-H. Jang, J.S. Ahn, H. Oh: Protuboxepins A and B and protubonines A and B from the marine-derived fungus Aspergillus sp. SF-5044, J. Nat. Prod. 74, 1284–1287 (2011)

    Article  CAS  Google Scholar 

  97. Y. Sun, K. Takada, Y. Takemoto, M. Yoshida, Y. Nogi, S. Okada, S. Matsunaga: Gliotoxin analogues from a marine-derived fungus, Penicillium sp., and their cytotoxic and histone methyltransferase inhibitory activities, J. Nat. Prod. 75, 111–114 (2012)

    Article  CAS  Google Scholar 

  98. B.S. Antia, T. Aree, C. Kasettrathat, S. Wiyakrutta, O.D. Ekpa, U.J. Ekpe, C. Mahidol, S. Ruchirawat, P. Kittakoop: Itaconic acid derivatives and diketopiperazine from the marine-derived fungus Aspergillus aculeatus CRI322-03, Phytochemistry 72, 816–820 (2011)

    Article  CAS  Google Scholar 

  99. O.I. Zhuravleva, S.S. Afiyatullov, V.A. Denisenko, S.P. Ermakova, N.N. Slinkina, P.S. Dmitrenok, N.Y. Kim: Secondary metabolites from a marine-derived fungus Aspergillus carneus Blochwitz, Phytochemistry 80, 123–131 (2012)

    Article  CAS  Google Scholar 

  100. S. Cai, X. Kong, W. Wang, H. Zhou, T. Zhu, D. Li, Q. Gu: Aspergilazine A, a diketopiperazine dimer with a rare N-1 to C-6 linkage, from a marine-derived fungus Aspergillus taichungensis, Tetrahedron Lett. 53, 2615–2617 (2012)

    Article  CAS  Google Scholar 

  101. K. Trisuwan, V. Rukachaisirikul, S. Phongpaichit, S. Preedanon, J. Sakayaroj: Madiolide and pyrone derivatives from the sea fan-derived fungus Curvularia sp. PSU-F22, Arch. Pharmacal Res. 34, 709–714 (2012)

    Article  CAS  Google Scholar 

  102. H. Ren, W.-W. Liu: Nidurufin as a new cell cycle inhibitor from marine-derived fungus Penicillium flavidorsum SHK1-27, Arch. Pharmacal Res. 34, 901–905 (2011)

    Article  CAS  Google Scholar 

  103. N. Khamthong, V. Rukachaisirikul, K. Tadpetch, M. Kaewpet, S. Phongpaichit, S. Preedanon, J. Sakayaroj: Tetrahydroanthraquinone and xanthone derivatives from the marine-derived fungus Trichoderma aureoviride PSU-F95, Arch. Pharmacal Res. 35, 461–468 (2012)

    Article  CAS  Google Scholar 

  104. H. Wang, Z. Lu, H.-J. Qu, P. Liu, C. Miao, T. Zhu, J. Li, K. Hong, W. Zhu: Antimicrobial aflatoxins from the marine-derived fungus Aspergillus flavus 092008, Arch. Pharmacal Res. 35, 1387–1392 (2012)

    Article  CAS  Google Scholar 

  105. C. Almeida, S. Kehraus, M. Prudencio, G.M. König: Marilones A–C, phthalides from the sponge-derived fungus Stachylidium sp., Beil. J. Org. Chem. 7, 1636–1642 (2011)

    Article  CAS  Google Scholar 

  106. C.-L. Shao, C.-Y. Wang, M.-Y. Wei, Y.-C. Gu, Z.-G. She, P.-Y. Qian, Y.-C. Lin: Aspergilones A and B, two benzylazaphilones with an unprecedented carbon skeleton from the gorgonian-derived fungus Aspergillus sp., Bioorg. Med. Chem. Lett. 21, 690–693 (2011)

    Article  CAS  Google Scholar 

  107. Z. Chen, Z. Zheng, H. Huang, Y. Song, X. Zhang, J. Ma, B. Wang, C. Zhang, J. Ju: Penicacids A–C, three new mycophenolic acid derivatives and immunosuppressive activities from the marine-derived fungus Penicillium sp. SOF07, Bioorg. Med. Chem. Lett. 22, 3332–3335 (2012)

    Article  CAS  Google Scholar 

  108. Y. Zhang, X.-M. Li, B.-G. Wang: Anthraquinone derivatives produced by marine-derived Aspergillus versicolor EN-7, Biosci. Biotechnol. Biochem. 76, 1774–1776 (2012)

    Article  CAS  Google Scholar 

  109. J.H. Sohn, H. Oh: Protulactones A and B: Two new polyketides from the marine-derived fungus Aspergillus sp. SF-5044, Bull. Kor. Chem. Soc. 31, 1695–1698 (2010)

    Article  CAS  Google Scholar 

  110. J.X. Yang, Y. Chen, C. Huang, Z. She, Y. Lin: A new isochroman derivative from the marine fungus Phomopsis sp. (No. ZH-111), Chem. Nat. Comp. 47, 13–16 (2011)

    Article  CAS  Google Scholar 

  111. C. Li, J. Zhang, C. Shao, W. Ding, Z. She, Y. Lin: A new xanthone derivative from the co-culture broth of two marine fungi (Strain No. E33 and K38), Chem. Nat. Comp. 47, 382–384 (2011)

    Article  CAS  Google Scholar 

  112. X. Xu, S. Zhao, J. Wei, N. Fang, L. Yin, J. Sun: Porric acid D from marine-derived fungus Alternaria sp. isolated from Bohai Sea, Chem. Nat. Comp. 47, 893–895 (2011)

    Article  CAS  Google Scholar 

  113. S. Li, M. Wei, G. Chen, Y. Lin: Two new dihydroisocoumarins from the endophytic fungus Aspergillus sp. collected from the South China Sea, Chem. Nat. Comp. 48, 371–373 (2012)

    Article  CAS  Google Scholar 

  114. L. Chen, W. Liu, X. Hu, K. Huang, J.-L. Wu, Q.-Q. Zhang: Citrinin derivatives from the marine-derived fungus Penicillium citrinum, Chem. Pharm. Bull. 59, 515–517 (2011)

    Article  CAS  Google Scholar 

  115. S. Cai, T. Zhu, L. Du , B. Zhao, D. Li, Q. Gu: Sterigmatocystins from the deep-sea-derived fungus Aspergillus versicolor, J. Antibiot. 64, 193–194 (2011)

    Article  CAS  Google Scholar 

  116. M.-Y. Kim, J.-H. Sohn, J.-H. Jang, J.S. Ahn, H. Oh: Two new botcinin derivatives encountered in the studies of secondary metabolites from the marine-derived fungus Botryotinia sp. SF5275, J. Antibiot. 65, 161–164 (2012)

    Article  CAS  Google Scholar 

  117. E. Julianti, H. Oh, K.H. Jang, J.K. Lee, S.K. Lee, D.-C. Oh, K.-B. Oh, J. Shin: Acremostrictin, a highly oxygenated metabolite from the marine fungus Acremonium strictum, J. Nat. Prod. 74, 2592–2594 (2011)

    Article  CAS  Google Scholar 

  118. D. Liu, X.-M. Li, L. Meng, C.-S. Li, S.-S. Gao, Z. Shang, P. Proksch, C.-G. Huang, B.-G. Wang: Nigerapyrones A–H, α-pyrone derivatives from the marine mangrove-derived endophytic fungus Aspergillus niger MA-132, J. Nat. Prod. 74, 1787–1791 (2011)

    Article  CAS  Google Scholar 

  119. J. Liu, F. Li, E.L. Kim, J.L. Li, J. Hong, K.S. Bae, H.Y. Chung, H.S. Kim, J.H. Jung: Antibacterial polyketides from the Jellyfish-derived fungus Paecilomyces variotii, J. Nat. Prod. 74, 1826–1829 (2011)

    Article  CAS  Google Scholar 

  120. C.-J. Zheng, C.-L. Shao, Z.-Y. Guo, J.-F. Chen, D.-S. Deng, K.-L. Yang, Y.-Y. Chen, X.-M. Fu, Z.-G. She, Y.-C. Lin, C.-Y. Wang: Bioactive hydroanthraquinones and anthraquinone dimers from a soft coral-derived Alternaria sp. fungus, J. Nat. Prod. 75, 189–197 (2012)

    Article  CAS  Google Scholar 

  121. Y. Myobatake, T. Takeuchi, K. Kuramochi, I. Kuriyama, T. Ishido, K. Hirano, F. Sugawara, H. Yoshida, Y. Mizushina: Pinophilins A and B, inhibitors of mammalian A-, B-, and Y-family DNA polymerases and human cancer cell proliferation, J. Nat. Prod. 75, 135–141 (2012)

    Article  CAS  Google Scholar 

  122. V. Rukachaisirikul, A. Rodglin, Y. Sukpondma, S. Phongpaichit, J. Buatong, J. Sakayaroj: Phthalide and isocoumarin derivatives produced by an Acremonium sp. isolated from a mangrove Rhizophora apiculata, J. Nat. Prod. 75, 853–858 (2012)

    Article  CAS  Google Scholar 

  123. K.-L. Yang, M.-Y. Wei, C.-L. Shao, X.-M. Fu, Z.-Y. Guo, R.-F. Xu, C.-J. Zheng, Z.-G. She, Y.-C. Lin, C.-Y. Wang: Antibacterial anthraquinone derivatives from a sea anemone-derived fungus Nigrospora sp., J. Nat. Prod. 75, 935–941 (2012)

    Article  CAS  Google Scholar 

  124. H. Huang, F. Wang, M. Luo, Y. Chen, Y. Song, W. Zhang, S. Zhang, J. Ju: Halogenated anthraquinones from the marine-derived fungus Aspergillus sp. SCSIO FO63, J. Nat. Prod. 75, 1346–1352 (2012)

    Article  CAS  Google Scholar 

  125. C.-H. Huang, J.-H. Pan, B. Chen, M. Yu, H.-B. Huang, X. Zhu, Y.-J. Lu, Z.-G. She, Y.-C. Lin: Three bianthraquinone derivatives from the mangrove endophytic fungus Alternaria sp. ZJ9-6B from the South China Sea, Mar. Drugs 9, 832–843 (2011)

    Article  CAS  Google Scholar 

  126. O.F. Smetanina, A.N. Yurchenko, S.S. Afiyatullov, A.I. Kalinovsky, M.A. Pushilin, Y.V. Khudyakova, N.N. Slinkina, S.P. Ermakova, E.A. Yurchenko: Oxirapentyns B–D produced by a marine-sediment fungus Isaria felina (DC.) Fr., Phytochem. Lett. 5, 165–169 (2012)

    Article  CAS  Google Scholar 

  127. G.F. Zhang, W.B. Han, J.T. Cui, S.W. Ng, Z.K. Guo, R.X. Tan, H.M. Ge: Neuraminidase inhibitory polyketides from the marine-derived fungus Phoma herbarum, Planta Medica 78, 76–78 (2012)

    Article  CAS  Google Scholar 

  128. L. Chen, T. Zhu, Y. Ding, I.A. Khan, Q. Gu, D. Li: Sorbiterrin A, a novel sorbicillin derivative with cholinesterase inhibition activity from the marine-derived fungus Penicillium terrestre, Tetrahedron Lett. 53, 325–328 (2012)

    Article  CAS  Google Scholar 

  129. D. Li, L. Chen, T. Zhu, T. Kurtán, A. Mándi, Z. Zhao, J. Li, Q. Gu: Chloctanspirones A and B, novel chlorinated polyketides with an unprecedented skeleton, from marine sediment derived fungus Penicillium terrestre, Tetrahedron 67, 7913–7918 (2011)

    Article  CAS  Google Scholar 

  130. N. Khamthong, V. Rukachaisirikul, S. Phongpaichit, S. Preedanon, J. Sakayaroj: Bioactive polyketides from the sea fan-derived fungus Penicillium citrinum, Tetrahedron 68, 8245–8250 (2012)

    Article  CAS  Google Scholar 

  131. M.F. Elsebai, S. Kehraus, U. Lindequist, F. Sasse, S. Shaaban, M. Gütschow, M. Josten, H.-G. Sahl, G.M. König: Antimicrobial phenalenone derivatives from the marine-derived fungus Coniothyrium cereal, Org. Biomol. Chem. 9, 802–808 (2011)

    Article  CAS  Google Scholar 

  132. W. Ebrahim, A.H. Aly, A. Mándi, F. Totzke, M.H.G. Kubbutat, V. Wray, W.-H. Lin, H. Dai, P. Proksch, T. Kurtán, A. Debbab: Decalactone derivatives from Corynespora cassicola, an endophytic fungus of the mangrove plant Laguncularia racemosa, Eur. J. Org. Chem. 2012, 3476–3484 (2012)

    Article  CAS  Google Scholar 

  133. D. Li, Y. Xu, C.-L. Shao, R.-Y. Yang, C.-J. Zheng, Y.-Y. Chen, X.-M. Fu, P.-Y. Qian, Z.-G. She, N.J. de Voogd, C.-Y. Wang: Antibacterial bisabolane-type sesquiterpenoids from the sponge-derived fungus Aspergillus sp., Mar. Drugs 10, 234–241 (2012)

    Article  CAS  Google Scholar 

  134. L.-L. Sun, C.-L. Shao, J.-F. Chen, Z.-Y. Guo, X.-M. Fu, M. Chen, Y.-Y. Chen, R. Li, N.J. de Voogd, Z.-G. She, Y.-C. Lin, C.-Y. Wang: New bisabolane sesquiterpenoids from a marine-derived fungus Aspergillus sp. isolated from the sponge Xestospongia testudinaria, Bioorg. Med. Chem. Lett. 22, 1326–1329 (2012)

    Article  CAS  Google Scholar 

  135. N. Ingavat, C. Mahidol, S. Ruchirawat, P. Kittakoop: Asperaculin A, a sesquiterpenoid from a marine-derived fungus, Aspergillus aculeatus, J. Nat. Prod. 74, 1650–1652 (2011)

    Article  CAS  Google Scholar 

  136. T. Kawahara, M. Takagi, K. Shin-ya: JBIR-124: A novel antioxidative agent from a marine-derived fungus Penicillium citrinum SpI080624G1f01, J. Antibiot. 65, 45–47 (2012)

    Article  CAS  Google Scholar 

  137. S.-S. Gao, X.-M. Li, C.-S. Li, P. Proksch, B.-G. Wang: Penicisteroids A and B, antifungal, cytotoxic polyoxgenated steroids from the marine alga-derived endophytic fungus Penicillium chrysogenum QEN-24S, Bioorg. Med. Chem. Lett. 21, 2894–2897 (2011)

    Article  CAS  Google Scholar 

  138. X. Xia, J. Zhang, Y. Zhang, F. Wei, X. Liu, A. Jia, C. Liu, W. Li, Z. She, Y. Lin: Pimarane diterpenes from the fungus Epicoccum sp. HS-1 associated with Apostichopus japonicas, Bioorg. Med. Chem. Lett. 22, 3017–3019 (2012)

    Article  CAS  Google Scholar 

  139. L. Sun, D. Li, M. Tao, Y. Chen, F. Dan, W. Zhang: Scopararanes C–G: new oxygenated pimarane diterpenes from the marine sediment-derived fungus Eutypella scoparia FS26, Mar. Drugs 10, 539–550 (2012)

    Article  CAS  Google Scholar 

  140. E.M.K. Wijeratne, B.P. Bashyal, M.X. Liu, D.D. Rocha, G.M.K.B. Gunaherath, J.M. U'Ren, M.K. Gunatilaka, A.E. Arnold, L. Whitesell, A.A.L. Gunatilaka: Geopyxins A–E, ent-kaurane diterpenoids from endolichenic fungal strains Geopyxis aff. majalis and Geopyxis sp. AZ0066: structure-activity relationships of geopyxins and their analogues, J. Nat. Prod. 75, 361–369 (2012)

    Article  CAS  Google Scholar 

  141. E. Cohen, L. Koch, K.M. Thu, Y. Rahamim, Y. Aluma, M. Ilan, O. Yarden, S. Carmeli: Novel terpenoids of the fungus Aspergillus insuetus isolated from the Mediterranean sponge Psammocinia sp. collected along the coast of Israel, Bioorg. Med. Chem. 19, 6587–6593 (2011)

    Article  CAS  Google Scholar 

  142. M. Arai, H. Niikawa, M. Kobayashi: Marine-derived fungal sesterterpenes, ophiobolins, inhibit biofilm formation of Mycobacterium species, J. Nat. Med. 67, 271–275 (2013)

    Article  CAS  Google Scholar 

  143. L.Y. Zang, W. Wei, Y. Guo, T. Wang, R.H. Jiao, S.W. Ng, R.X. Tan, H.M. Ge: Sesquiterpenoids from the mangrove-derived endophytic fungus Diaporthe sp., J. Nat. Prod. 75, 1744–1749 (2012)

    Article  CAS  Google Scholar 

  144. I.E. Mohamed, S. Kehraus, A. Krick, G.M. König, G. Kelter, A. Maier, H.-H. Fiebig, M. Kalesse, N.P. Malek, H. Gross: Mode of action of epoxyphomalins A and B and characterization of related metabolites from the marine-derived fungus Paraconiothyrium sp., J. Nat. Prod. 73, 2053–2056 (2010)

    Article  CAS  Google Scholar 

  145. A.-F. Sun, X.-M. Li, L. Meng, C.-M. Cui, S.-S. Gao, C.-S. Li, C.-G. Huang, B.-G. Wang: Asperolides A–C, tetranorlabdane diterpenoids from the marine alga-derived endophytic fungus Aspergillus wentii EN-48, J. Nat. Prod. 75, 148–152 (2012)

    Article  CAS  Google Scholar 

  146. Y. Zhou, A. Mándi, A. Debbab, V. Wray, B. Schulz, W.E.G. Müller, W. Lin, P. Proksch, T. Kurtán, A.H. Aly: New austalides from the sponge-associated fungus Aspergillus sp., Eur. J. Org. Chem. 2011, 6009–6019 (2011)

    Article  CAS  Google Scholar 

  147. W.-L. Mei, B. Zheng, Y.-X. Zhao, H.-M. Zhong, X.-L.W. Chen, Y.-B. Zeng, W.-H. Dong, J.-L. Huang, P. Proksch, H.-F. Dai: Meroterpenes from endophytic fungus A1 of mangrove plant Scyphiphora hydrophyllacea, Mar. Drugs 10, 1993–2001 (2012)

    Article  CAS  Google Scholar 

  148. L.-N. Zhou, H.-Q. Gao, S.-X. Cai, T.-J. Zhu, Q.-Q. Gu, D.-H. Li: Two new cyclic pentapeptides from the marine-derived fungus Aspergillus versicolor, Helv. Chim. Acta 94, 1065–1070 (2011)

    Article  CAS  Google Scholar 

  149. Z. Chen, Y. Song, Y. Chen, H. Huang, W. Zhang, J. Ju: Cyclic heptapeptides, cordyheptapeptides C–E, from the marine-derived fungus Acremonium persicinum SCSIO 115 and their cytotoxic activities, J. Nat. Prod. 75, 1215–1219 (2012)

    Article  CAS  Google Scholar 

  150. H.C. Vervoort, M. Draškovic, P. Crews: Histone deacetylase inhibitors as a tool to up-regulate new fungal biosynthetic products: Isolation of EGM-556, a cyclodepsipeptide, from Microascus sp., Org. Lett. 13, 410–413 (2011)

    Article  CAS  Google Scholar 

  151. Q.X. Wu, X.J. Jin, M. Draškovic, M.S. Crews, K. Tenney, F.A. Valeriote, X.J. Yao, P. Crews: Unraveling the numerous biosynthetic products of the marine sediment-derived fungus, Aspergillus insulicola, Phytochem. Lett. 5, 114–117 (2012)

    Article  CAS  Google Scholar 

  152. W. Ebrahim, J. Kjer, M. El Amrani, V. Wray, W. Lin, R. Ebel, D. Lai, P. Proksch: Pullularins E and F, two new peptides from the endophytic fungus Bionectria ochroleuca isolated form the mangrove plant Sonneratia caseolaris, Mar. Drugs 10, 1081–1091 (2012)

    Article  CAS  Google Scholar 

  153. S.W. Meyer, T.F. Mordhorst, C. Lee, P.R. Jensen, W. Fenical, M. Köck: Penilumamide, a novel lumazine peptide isolated from the marine-derived fungus, Penicillium sp., Org. Biomol. Chem. CNL-338(8), 2158–2163 (2010)

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sherif S. Ebada .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Springer-Verlag Berlin Heidelberg

About this chapter

Cite this chapter

Ebada, S.S., Proksch, P. (2015). Marine-Derived Fungal Metabolites. In: Kim, SK. (eds) Springer Handbook of Marine Biotechnology. Springer Handbooks. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-642-53971-8_32

Download citation

Publish with us

Policies and ethics