메뉴 건너뛰기




Volumn 8, Issue 1, 2009, Pages 69-85

Drug development from marine natural products

Author keywords

[No Author keywords available]

Indexed keywords

ALKALOID; ANTHRAMYCIN; ARENASTATIN A; BETA NAPHTHOFLAVONE; CRYPTOPHYCIN 52; DEHYDRODIDEMNIN B; DEXAMETHASONE; DIDEMNIN B; DISCODERMOLIDE; DOLASTATIN 10; DOXORUBICIN; ET 729; HALICHONDRIN B; IRINOTECAN; KAHALALIDE F; MORPHINE; NATURAL PRODUCT; OKADAIC ACID; OMEGA CONOTOXIN MVIIA; PACLITAXEL; PHOMOPSIN A; PLATINUM COMPLEX; SAFRAMYCIN A; SOBLIDOTIN; TALTOBULIN; TRABECTEDIN; UNCLASSIFIED DRUG; UNINDEXED DRUG; VINBLASTINE; VINCRISTINE; XESTOSPONGIN C;

EID: 58149189779     PISSN: 14741776     EISSN: 14741784     Source Type: Journal    
DOI: 10.1038/nrd2487     Document Type: Review
Times cited : (1008)

References (287)
  • 2
    • 4344650390 scopus 로고    scopus 로고
    • Marine natural products and related compounds in clinical and advanced preclinical trials
    • Newman D. J. & Cragg G. M. Marine natural products and related compounds in clinical and advanced preclinical trials. J. Nat. Prod. 67 1216-1238 (2004).
    • (2004) J. Nat. Prod , vol.67 , pp. 1216-1238
    • Newman, D.J.1    Cragg, G.M.2
  • 3
    • 34247109045 scopus 로고    scopus 로고
    • Natural products as sources of new drugs over the last 25 years
    • Newman D. J. & Cragg G. M. Natural products as sources of new drugs over the last 25 years. J. Nat. Prod. 70, 461-477 (2007).
    • (2007) J. Nat. Prod , vol.70 , pp. 461-477
    • Newman, D.J.1    Cragg, G.M.2
  • 5
    • 0000719402 scopus 로고
    • Contributions to the study of marine products. XXXII. The nucleosides of sponges. I
    • Bergmann, W. & Feeney R. J. Contributions to the study of marine products. XXXII. The nucleosides of sponges. I. J. Org. Chem. 16, 981-987 (1951).
    • (1951) J. Org. Chem , vol.16 , pp. 981-987
    • Bergmann, W.1    Feeney, R.J.2
  • 6
    • 0000833362 scopus 로고
    • Contributions to the study of marine products. XL. The nucleosides of sponges. IV. Spongosine
    • Bergmann, W. & Burke, D. C. Contributions to the study of marine products. XL. The nucleosides of sponges. IV. Spongosine. J. Org. Chem. 22, 226-228 (1956).
    • (1956) J. Org. Chem , vol.22 , pp. 226-228
    • Bergmann, W.1    Burke, D.C.2
  • 7
    • 33947463084 scopus 로고
    • Contributions to the study of marine products. XLIII. The nucleosides of sponges. V. The synthesis of spongosine
    • Bergmann, W. & Stempien, M. F. Contributions to the study of marine products. XLIII. The nucleosides of sponges. V. The synthesis of spongosine. J. Org. Chem. 22, 1575-1557 (1957).
    • (1957) J. Org. Chem , vol.22 , pp. 1575-1557
    • Bergmann, W.1    Stempien, M.F.2
  • 8
    • 14944383798 scopus 로고    scopus 로고
    • The evolving role of natural products in drug discovery
    • Koehn, F. E. & Carter, G. T. The evolving role of natural products in drug discovery. Nature Rev. Drug Discovery 4, 206-220 (2004).
    • (2004) Nature Rev. Drug Discovery , vol.4 , pp. 206-220
    • Koehn, F.E.1    Carter, G.T.2
  • 9
    • 35448976394 scopus 로고    scopus 로고
    • Liquid gold mine
    • Jarvis, L. M. Liquid gold mine. Chem. Engin. News 85, 22-28 (2007).
    • (2007) Chem. Engin. News , vol.85 , pp. 22-28
    • Jarvis, L.M.1
  • 10
    • 0002829177 scopus 로고    scopus 로고
    • ed. Fusetani, N, Karger, Basel
    • Mendola, D. in Drugs from the Sea (ed. Fusetani, N.) 120-133 (Karger, Basel, 2000).
    • (2000) Drugs from the Sea , pp. 120-133
    • Mendola, D.1
  • 11
    • 0022373275 scopus 로고
    • Peptide neurotoxins from fish-hunting cone snails
    • Olivera, B. M. et al. Peptide neurotoxins from fish-hunting cone snails. Science 230, 1338-1343 (1985).
    • (1985) Science , vol.230 , pp. 1338-1343
    • Olivera, B.M.1
  • 12
    • 0347989461 scopus 로고    scopus 로고
    • Conus venoms: A rich source of novel ion channel-targeted peptides
    • Terlau, H. & Olivera, B. M. Conus venoms: A rich source of novel ion channel-targeted peptides. Physiol. Rev. 84, 41-68 (2004).
    • (2004) Physiol. Rev , vol.84 , pp. 41-68
    • Terlau, H.1    Olivera, B.M.2
  • 13
    • 0028292145 scopus 로고
    • Calcium-channel diversity and neurotransmitter release - the ω-conotoxins and ω-agatoxins
    • Olivera, B. M., Miljanich, G. P., Ramachandran, J. & Adams, M. E. Calcium-channel diversity and neurotransmitter release - the ω-conotoxins and ω-agatoxins. Ann. Rev. Biochem. 63, 823-867 (1994).
    • (1994) Ann. Rev. Biochem , vol.63 , pp. 823-867
    • Olivera, B.M.1    Miljanich, G.P.2    Ramachandran, J.3    Adams, M.E.4
  • 15
    • 0032493315 scopus 로고    scopus 로고
    • Roles of individual disulfide bonds in the stability and folding of an ω-conotoxin
    • Price-Carter, M., Hull, M. S. & Goldenberg, D. P. Roles of individual disulfide bonds in the stability and folding of an ω-conotoxin. Biochemistry 37, 9851-9861 (1998).
    • (1998) Biochemistry , vol.37 , pp. 9851-9861
    • Price-Carter, M.1    Hull, M.S.2    Goldenberg, D.P.3
  • 16
    • 0023187486 scopus 로고
    • Neuronal calcium-channel antagonists - discrimination between calcium-channel subtypes using ω-conotoxin from Conus magus venom
    • Olivera, B. M. et al. Neuronal calcium-channel antagonists - discrimination between calcium-channel subtypes using ω-conotoxin from Conus magus venom. Biochemistry 26, 2086-2090 (1987).
    • (1987) Biochemistry , vol.26 , pp. 2086-2090
    • Olivera, B.M.1
  • 17
    • 0040040615 scopus 로고
    • Transmitter release from presynaptic terminals of electric organ: Inhibition by the calcium channel antagonist ω Conus toxin
    • Yeager, R. E., Yoshikami, D., Rivier, J., Cruz, L. J. & Miljanich, G. P. Transmitter release from presynaptic terminals of electric organ: inhibition by the calcium channel antagonist ω Conus toxin. J. Neurosci. 7, 2390-2396 (1987).
    • (1987) J. Neurosci , vol.7 , pp. 2390-2396
    • Yeager, R.E.1    Yoshikami, D.2    Rivier, J.3    Cruz, L.J.4    Miljanich, G.P.5
  • 18
    • 0034636131 scopus 로고    scopus 로고
    • Structural and dynamic characterization of ω -conotoxin MVIIA: The binding loop exhibits slow conformational exchange
    • Atkinson, R. A., Kieffer, B., Dejaegere, A., Sirockin, F. & Lefevre, J. F. Structural and dynamic characterization of ω -conotoxin MVIIA: The binding loop exhibits slow conformational exchange. Biochemistry 39, 3908-3919 (2000).
    • (2000) Biochemistry , vol.39 , pp. 3908-3919
    • Atkinson, R.A.1    Kieffer, B.2    Dejaegere, A.3    Sirockin, F.4    Lefevre, J.F.5
  • 19
    • 0028985407 scopus 로고
    • Tyr13 Is essential for the activity of ω -conotoxin MVIIA and GVIA, specific N-type calcium channel blockers
    • Kim, J. I., Takahashi, M., Ohtake, A., Wakamiya, A. & Sato, K. Tyr13 Is essential for the activity of ω -conotoxin MVIIA and GVIA, specific N-type calcium channel blockers. Biochem. Biophys. Res. Commun. 206, 449-454 (1995).
    • (1995) Biochem. Biophys. Res. Commun , vol.206 , pp. 449-454
    • Kim, J.I.1    Takahashi, M.2    Ohtake, A.3    Wakamiya, A.4    Sato, K.5
  • 20
    • 0028105906 scopus 로고
    • Neuroanatomical distribution of receptors for a novel voltage-sensitive calcium-channel antagonist, SNX-230 (ω -conopeptide MVIIC)
    • Gohil, K., Bell, J. R., Ramachandran, J. & Miljanich, G. P. Neuroanatomical distribution of receptors for a novel voltage-sensitive calcium-channel antagonist, SNX-230 (ω -conopeptide MVIIC). Brain Res. 653, 258-266 (1994).
    • (1994) Brain Res , vol.653 , pp. 258-266
    • Gohil, K.1    Bell, J.R.2    Ramachandran, J.3    Miljanich, G.P.4
  • 21
    • 0030434482 scopus 로고    scopus 로고
    • Selective N-type neuronal voltage-sensitive calcium channel blocker, SNX-111, produces spinal antinociception in rat models of acute, persistent and neuropathic pain
    • Bowersox, S. S. et al. Selective N-type neuronal voltage-sensitive calcium channel blocker, SNX-111, produces spinal antinociception in rat models of acute, persistent and neuropathic pain. J. Pharmacol. Exp. Ther. 279, 1243-1249 (1996).
    • (1996) J. Pharmacol. Exp. Ther , vol.279 , pp. 1243-1249
    • Bowersox, S.S.1
  • 22
    • 0028303526 scopus 로고
    • Characterization of the binding of ω -conopeptides to different classes of non-L-type neuronal calcium channels
    • Kristipati, R. et al. Characterization of the binding of ω -conopeptides to different classes of non-L-type neuronal calcium channels. Mol. Cell. Neurosci. 5, 219-228 (1994).
    • (1994) Mol. Cell. Neurosci , vol.5 , pp. 219-228
    • Kristipati, R.1
  • 23
    • 0035065039 scopus 로고    scopus 로고
    • Jones, R. M. et al. Composition and therapeutic utility of conotoxins from genus Conus. Patent status 1996-2000 Exp. Opin. Ther. Patents 11, 603-623 2001
    • Jones, R. M. et al. Composition and therapeutic utility of conotoxins from genus Conus. Patent status 1996-2000 Exp. Opin. Ther. Patents 11, 603-623 (2001).
  • 24
    • 0031796866 scopus 로고    scopus 로고
    • Pharmacotherapeutic potential of ω-conotoxin MVIIA (SNX-111), an N-type neuronal calcium channel blocker found in the venom of Conus magus
    • Bowersox, S. S. & Luther, R. Pharmacotherapeutic potential of ω-conotoxin MVIIA (SNX-111), an N-type neuronal calcium channel blocker found in the venom of Conus magus. Toxicon 36, 1651-1658 (1998).
    • (1998) Toxicon , vol.36 , pp. 1651-1658
    • Bowersox, S.S.1    Luther, R.2
  • 25
    • 0033963578 scopus 로고    scopus 로고
    • Effects of intrathecal administration of ziconotide, a selective neuronal N-type calcium channel blocker, on mechanical allodynia and heat hyperalgesia in a rat model of postoperative pain
    • Wang, Y. X., Pettus, M., Gao, D., Phillips, C. & Scott Bowersox, S. Effects of intrathecal administration of ziconotide, a selective neuronal N-type calcium channel blocker, on mechanical allodynia and heat hyperalgesia in a rat model of postoperative pain. Pain 84 151-158 (2000).
    • (2000) Pain , vol.84 , pp. 151-158
    • Wang, Y.X.1    Pettus, M.2    Gao, D.3    Phillips, C.4    Scott Bowersox, S.5
  • 26
    • 9144272640 scopus 로고    scopus 로고
    • Neuronal calcium channel blocker for treating severe chronic pain
    • Miljanich, G. P. Ziconotide: Neuronal calcium channel blocker for treating severe chronic pain. Curr. Med. Chem. 11, 3029-3040 (2004).
    • (2004) Curr. Med. Chem , vol.11 , pp. 3029-3040
    • Miljanich, G.P.Z.1
  • 27
    • 23644456345 scopus 로고    scopus 로고
    • Peptide leads new class of chronic pain drugs
    • Garber, K. Peptide leads new class of chronic pain drugs. Nature Biotech. 23, 399 (2005).
    • (2005) Nature Biotech , vol.23 , pp. 399
    • Garber, K.1
  • 28
    • 9944260795 scopus 로고    scopus 로고
    • Conus toxins: Targets and properties
    • Heading, C. E. Conus toxins: Targets and properties. IDrugs 7, 1011-1016 (2004).
    • (2004) IDrugs , vol.7 , pp. 1011-1016
    • Heading, C.E.1
  • 29
    • 58149177612 scopus 로고    scopus 로고
    • Sigel, M. M. et al. in Food-Drugs from the Sea: Proceedings (ed. Youngken, H. W. Jr) 281-294 (Marine Technology Society, Washington, DC, 1969).
    • Sigel, M. M. et al. in Food-Drugs from the Sea: Proceedings (ed. Youngken, H. W. Jr) 281-294 (Marine Technology Society, Washington, DC, 1969).
  • 30
    • 0025070774 scopus 로고
    • Ecteinascidin-729, Ecteinascidin-743, Ecteinascidin-745, Ecteinascidin-759a, Ecteinascidin-759b, and Ecteinascidin-770 - potent antitumor agents from the Caribbean tunicate Ecteinascidia turbinata
    • Rinehart, K. L. et al. Ecteinascidin-729, Ecteinascidin-743, Ecteinascidin-745, Ecteinascidin-759a, Ecteinascidin-759b, and Ecteinascidin-770 - potent antitumor agents from the Caribbean tunicate Ecteinascidia turbinata. J. Org. Chem. 55, 4512-4515 (1990).
    • (1990) J. Org. Chem , vol.55 , pp. 4512-4515
    • Rinehart, K.L.1
  • 31
    • 0024995421 scopus 로고
    • Antitumor tetrahyrodisoquinoline alkaloids from the colonial ascidian Ecteinascidia turbinata
    • Wright, A. E. et al. Antitumor tetrahyrodisoquinoline alkaloids from the colonial ascidian Ecteinascidia turbinata. J. Org. Chem. 55, 4508-4512 (1990).
    • (1990) J. Org. Chem , vol.55 , pp. 4508-4512
    • Wright, A.E.1
  • 32
    • 0026474576 scopus 로고
    • Additional antitumor ecteinascidins from a Caribbean tunicate: Crystal structures and activities in vivo
    • Sakai, R., Rinehart, K. L., Guan, Y. & Wang, A. H. Additional antitumor ecteinascidins from a Caribbean tunicate: Crystal structures and activities in vivo. Proc. Nat. Acad. Sci. USA 89, 11456-11460 (1992).
    • (1992) Proc. Nat. Acad. Sci. USA , vol.89 , pp. 11456-11460
    • Sakai, R.1    Rinehart, K.L.2    Guan, Y.3    Wang, A.H.4
  • 33
    • 0029805757 scopus 로고    scopus 로고
    • Enantioselective total synthesis of ecteinascidin 743
    • Corey, E. J., Gin, D. Y. & Kania, R. S. Enantioselective total synthesis of ecteinascidin 743. J. Am. Chem. Soc. 118, 9202-9203 (1996).
    • (1996) J. Am. Chem. Soc , vol.118 , pp. 9202-9203
    • Corey, E.J.1    Gin, D.Y.2    Kania, R.S.3
  • 34
    • 0034611483 scopus 로고    scopus 로고
    • A new, more efficient, and effective process for the synthesis of a key pentacyclic intermediate for production of ecteinascidin and phthalascidin antitumor agents
    • Martinez, E. J. & Corey, E. J. A new, more efficient, and effective process for the synthesis of a key pentacyclic intermediate for production of ecteinascidin and phthalascidin antitumor agents. Org. Lett. 2, 993-996 (2000).
    • (2000) Org. Lett , vol.2 , pp. 993-996
    • Martinez, E.J.1    Corey, E.J.2
  • 35
    • 0034632436 scopus 로고    scopus 로고
    • Synthesis of ecteinascidin ET-743 and Phthalascidin Pt-650 from cyanosafracin B
    • Cuevas, C. et al. Synthesis of ecteinascidin ET-743 and Phthalascidin Pt-650 from cyanosafracin B. Org. Lett. 2, 2545-2548 (2000).
    • (2000) Org. Lett , vol.2 , pp. 2545-2548
    • Cuevas, C.1
  • 36
    • 0036021010 scopus 로고    scopus 로고
    • Ecteinascidin 743: A novel anticancer drug with a unique mechanism of action
    • Aune, G. J., Furuta, T. & Pommier, Y. Ecteinascidin 743: A novel anticancer drug with a unique mechanism of action. Anticancer Drugs 13, 545-555 (2002).
    • (2002) Anticancer Drugs , vol.13 , pp. 545-555
    • Aune, G.J.1    Furuta, T.2    Pommier, Y.3
  • 37
    • 0029838307 scopus 로고    scopus 로고
    • DNA sequence- and structure-selective alkylation of guanine N2 in the DNA minor groove by ecteinascidin 743, a potent antitumor compound from the Caribbean tunicate Ecteinascidia turbinata
    • Pommier, Y. et al. DNA sequence- and structure-selective alkylation of guanine N2 in the DNA minor groove by ecteinascidin 743, a potent antitumor compound from the Caribbean tunicate Ecteinascidia turbinata. Biochemistry 35, 13303-13309 (1996).
    • (1996) Biochemistry , vol.35 , pp. 13303-13309
    • Pommier, Y.1
  • 38
    • 0033566150 scopus 로고    scopus 로고
    • Ecteinascidin 743: A minor groove alkylator that bends DNA toward the major groove
    • Zewail-Foote, M. & Hurley, L. H. Ecteinascidin 743: A minor groove alkylator that bends DNA toward the major groove. J. Med. Chem. 42, 2493-2497 (1999).
    • (1999) J. Med. Chem , vol.42 , pp. 2493-2497
    • Zewail-Foote, M.1    Hurley, L.H.2
  • 39
    • 0035199322 scopus 로고    scopus 로고
    • The inefficiency of incisions of ecteinascidin 743-DNA adducts by the UvrABC nuclease and the unique structural feature of the DNA adducts can be used to explain the repair-dependent toxicities of this antitumor agent
    • Zewail-Foote, M. et al. The inefficiency of incisions of ecteinascidin 743-DNA adducts by the UvrABC nuclease and the unique structural feature of the DNA adducts can be used to explain the repair-dependent toxicities of this antitumor agent. Chem. Biol. 8, 1033-1049 (2001).
    • (2001) Chem. Biol , vol.8 , pp. 1033-1049
    • Zewail-Foote, M.1
  • 40
    • 17944374027 scopus 로고    scopus 로고
    • Antiproliferative activity of ecteinascidin 743 is dependent upon transcription-coupled nucleotide-excision repair
    • Takebayashi, Y. et al. Antiproliferative activity of ecteinascidin 743 is dependent upon transcription-coupled nucleotide-excision repair. Nature Med. 7, 961-966 (2001).
    • (2001) Nature Med , vol.7 , pp. 961-966
    • Takebayashi, Y.1
  • 41
    • 58149185533 scopus 로고    scopus 로고
    • Final results of a phase II trial of 3-HR infusion trabectedin in patients with recurrent sarcomas
    • Di Leo, P. et al. Final results of a phase II trial of 3-HR infusion trabectedin in patients with recurrent sarcomas. Ann. Oncol. 17, 167-167 (2006).
    • (2006) Ann. Oncol , vol.17 , pp. 167-167
    • Di Leo, P.1
  • 42
    • 34250329180 scopus 로고    scopus 로고
    • Phase II study of trabectedin in pretreated patients with advanced colorectal cancer
    • Paz-Ares, L. et al. Phase II study of trabectedin in pretreated patients with advanced colorectal cancer. Clin. Colorectal Cancer 6, 522-528 (2007).
    • (2007) Clin. Colorectal Cancer , vol.6 , pp. 522-528
    • Paz-Ares, L.1
  • 43
    • 33745259879 scopus 로고    scopus 로고
    • Zelek, L. et al. A phase II study of Yondelis® (trabectedin, ET-743) as a 24-h continuous intravenous infusion in pretreated advanced breast cancer Br. J. Cancer 94, 1610-1614 (2006).
    • Zelek, L. et al. A phase II study of Yondelis® (trabectedin, ET-743) as a 24-h continuous intravenous infusion in pretreated advanced breast cancer Br. J. Cancer 94, 1610-1614 (2006).
  • 44
    • 20144377216 scopus 로고    scopus 로고
    • Trabectedin for women with ovarian carcinoma after treatment with platinum and taxanes fails
    • Sessa, C. et al. Trabectedin for women with ovarian carcinoma after treatment with platinum and taxanes fails. J. Clin. Oncol. 23, 1867-1874 (2005).
    • (2005) J. Clin. Oncol , vol.23 , pp. 1867-1874
    • Sessa, C.1
  • 45
    • 1542398698 scopus 로고    scopus 로고
    • Phase II study of ecteinascidin-743 in advanced pretreated soft tissue sarcoma patients
    • Yovine, A. et al. Phase II study of ecteinascidin-743 in advanced pretreated soft tissue sarcoma patients. J. Clin. Oncol. 22, 890-899 (2004).
    • (2004) J. Clin. Oncol , vol.22 , pp. 890-899
    • Yovine, A.1
  • 46
    • 0242522296 scopus 로고    scopus 로고
    • Therapeutic impact of ET-743 (Yondelis; trabectidin), a new marine-derived compound, in sarcoma
    • Jimeno, J. et al. Therapeutic impact of ET-743 (Yondelis; trabectidin), a new marine-derived compound, in sarcoma. Curr. Op. Orthopaed. 14, 419-428 (2003).
    • (2003) Curr. Op. Orthopaed , vol.14 , pp. 419-428
    • Jimeno, J.1
  • 47
    • 1942452836 scopus 로고    scopus 로고
    • Progress in the clinical development of new marine-derived anticancer compounds
    • Jimeno, J. et al. Progress in the clinical development of new marine-derived anticancer compounds. Anticancer Drugs 15, 321-329 (2004).
    • (2004) Anticancer Drugs , vol.15 , pp. 321-329
    • Jimeno, J.1
  • 48
    • 0036913513 scopus 로고    scopus 로고
    • A phase II and pharmacokinetic study of ecteinascidin 743 in patients with gastrointestinal stromal tumors
    • Ryan, D. P. et al. A phase II and pharmacokinetic study of ecteinascidin 743 in patients with gastrointestinal stromal tumors. Oncologist 7, 531-538 (2002).
    • (2002) Oncologist , vol.7 , pp. 531-538
    • Ryan, D.P.1
  • 49
    • 2942571277 scopus 로고    scopus 로고
    • A phase II study of ET-743/trabectedin ('Yondelis') for patients with advanced gastrointestinal stromal tumours
    • Blay, J. Y. et al. A phase II study of ET-743/trabectedin ('Yondelis') for patients with advanced gastrointestinal stromal tumours. Eur. J. Cancer 40, 1327-1331 (2004).
    • (2004) Eur. J. Cancer , vol.40 , pp. 1327-1331
    • Blay, J.Y.1
  • 50
    • 0042878545 scopus 로고    scopus 로고
    • Effective combination of ET-743 and doxorubicin in sarcoma: Preclinical studies
    • Meco, D. et al. Effective combination of ET-743 and doxorubicin in sarcoma: Preclinical studies. Cancer Chemother. Pharmacol. 52 131-138 (2003).
    • (2003) Cancer Chemother. Pharmacol , vol.52 , pp. 131-138
    • Meco, D.1
  • 51
    • 0034786774 scopus 로고    scopus 로고
    • Sequence-dependent enhancement of cytotoxicity produced by ecteinascidin 743 (ET-743) with doxorubicin or paclitaxel in soft tissue sarcoma cells
    • Takahashi, N., Li, W. W., Banerjee, D., Scotto, K. W. & Bertino, J. R. Sequence-dependent enhancement of cytotoxicity produced by ecteinascidin 743 (ET-743) with doxorubicin or paclitaxel in soft tissue sarcoma cells. Clin. Canc. Res. 7, 3251-3257 (2001).
    • (2001) Clin. Canc. Res , vol.7 , pp. 3251-3257
    • Takahashi, N.1    Li, W.W.2    Banerjee, D.3    Scotto, K.W.4    Bertino, J.R.5
  • 52
    • 24044480041 scopus 로고    scopus 로고
    • Combination of trabectedin and irinotecan is highly effective in a human rhabdomyosarcoma xenograft
    • Riccardi, A. et al. Combination of trabectedin and irinotecan is highly effective in a human rhabdomyosarcoma xenograft. Anticancer Drugs 16, 811-815 (2005).
    • (2005) Anticancer Drugs , vol.16 , pp. 811-815
    • Riccardi, A.1
  • 53
    • 26244458402 scopus 로고    scopus 로고
    • In-vitro cytotoxicity of ET-743 (Trabectedin, Yondelis), a marine anti-cancer drug, in the Hep G2 cell line: Influence of cytochrome P450 and phase II inhibition, and cytochrome P450 induction
    • Brandon, E. F. A. et al. In-vitro cytotoxicity of ET-743 (Trabectedin, Yondelis), a marine anti-cancer drug, in the Hep G2 cell line: Influence of cytochrome P450 and phase II inhibition, and cytochrome P450 induction. Anticancer Drugs 16, 935-943 (2005).
    • (2005) Anticancer Drugs , vol.16 , pp. 935-943
    • Brandon, E.F.A.1
  • 54
    • 1642388840 scopus 로고    scopus 로고
    • Comparison of four modulators of drug metabolism as protectants against the hepatotoxicity of the novel antitumor drug yondelis (ET-743) in the female rat and in hepatocytes in vitro
    • Donald, S. et al. Comparison of four modulators of drug metabolism as protectants against the hepatotoxicity of the novel antitumor drug yondelis (ET-743) in the female rat and in hepatocytes in vitro. Cancer Chemother. Pharmacol. 53, 305-312 (2004).
    • (2004) Cancer Chemother. Pharmacol , vol.53 , pp. 305-312
    • Donald, S.1
  • 55
    • 0141731283 scopus 로고    scopus 로고
    • Complete protection by high-dose dexamethasone against the hepatotoxicity of the novel antitumor drug yondelis (ET-743) in the rat
    • Donald, S. et al. Complete protection by high-dose dexamethasone against the hepatotoxicity of the novel antitumor drug yondelis (ET-743) in the rat. Cancer Res. 63, 5902-5908 (2003).
    • (2003) Cancer Res , vol.63 , pp. 5902-5908
    • Donald, S.1
  • 56
    • 14644423300 scopus 로고    scopus 로고
    • Hepatotoxicity and metabolism of trabectedin: A literature review
    • Beumer, J. H., Schellens, J. H. & Beijnen, J. H. Hepatotoxicity and metabolism of trabectedin: A literature review. Pharmacol. Res. 51, 391-398 (2005).
    • (2005) Pharmacol. Res , vol.51 , pp. 391-398
    • Beumer, J.H.1    Schellens, J.H.2    Beijnen, J.H.3
  • 57
    • 0023026034 scopus 로고
    • Didemnin B. The first marine compound entering clinical trials as an antineoplastic agent
    • Chun, H. G. et al. Didemnin B. The first marine compound entering clinical trials as an antineoplastic agent. Invest. New Drugs 4 279-284 (1986).
    • (1986) Invest. New Drugs , vol.4 , pp. 279-284
    • Chun, H.G.1
  • 58
    • 0036178591 scopus 로고    scopus 로고
    • Natural products as probes of cell biology: 20 years of didemnin research
    • Vera, M. D. & Joullie, M. M. Natural products as probes of cell biology: 20 years of didemnin research. Med. Res. Rev. 22, 102-145 (2002).
    • (2002) Med. Res. Rev , vol.22 , pp. 102-145
    • Vera, M.D.1    Joullie, M.M.2
  • 59
    • 0033981511 scopus 로고    scopus 로고
    • Antitumor Compounds from Tunicates
    • Rinehart, K. Antitumor Compounds from Tunicates. Med. Res. Rev. 1, 1-27 (2003).
    • (2003) Med. Res. Rev , vol.1 , pp. 1-27
    • Rinehart, K.1
  • 60
    • 0019470678 scopus 로고
    • Structures of the didemnins, antiviral and cytotoxic depsipeptides from a Carribean tunicate
    • Rinehart, K. L. Jr., Gloer, J. B., Cook, J. C., Mizsak, S. A. & Scahill, T. A. Structures of the didemnins, antiviral and cytotoxic depsipeptides from a Carribean tunicate. J. Am. Chem. Soc. 103, 1857-1859 (1981).
    • (1981) J. Am. Chem. Soc , vol.103 , pp. 1857-1859
    • Rinehart Jr., K.L.1    Gloer, J.B.2    Cook, J.C.3    Mizsak, S.A.4    Scahill, T.A.5
  • 61
    • 0019469697 scopus 로고
    • Didemnins: Antiviral and antitumor depsipeptides from a caribbean tunicate
    • Rinehart, K. L. Jr et al. Didemnins: Antiviral and antitumor depsipeptides from a caribbean tunicate. Science 212, 933-935 (1981).
    • (1981) Science , vol.212 , pp. 933-935
    • Rinehart Jr, K.L.1
  • 62
    • 0001686304 scopus 로고
    • Total synthesis of didemnins A, B, and C
    • Rinehart, K. L. Jr et al. Total synthesis of didemnins A, B, and C. J. Am. Chem. Soc. 109, 6856-6848 (1987).
    • (1987) J. Am. Chem. Soc , vol.109 , pp. 6856-6848
    • Rinehart Jr, K.L.1
  • 63
    • 0024328232 scopus 로고
    • The complete spectral assignment of didemnin B and nordidemnin B
    • McKee, T. C., Ireland, C. M., Lindquist, N. & Fenical, W. The complete spectral assignment of didemnin B and nordidemnin B. Tetrahedron Lett. 30, 3053-3056 (1989).
    • (1989) Tetrahedron Lett , vol.30 , pp. 3053-3056
    • McKee, T.C.1    Ireland, C.M.2    Lindquist, N.3    Fenical, W.4
  • 65
    • 0000672058 scopus 로고
    • Crystal and molecular structure of didemnin B, an antiviral and cytotoxic depsipeptide
    • Hossain, M. B. et al. Crystal and molecular structure of didemnin B, an antiviral and cytotoxic depsipeptide. Proc. Natl Acad. Sci. USA 85, 4118-4122 (1988).
    • (1988) Proc. Natl Acad. Sci. USA , vol.85 , pp. 4118-4122
    • Hossain, M.B.1
  • 66
    • 0032555255 scopus 로고    scopus 로고
    • The antiproliferative agent didemnin B uncompetitively inhibits palmitoyl protein thioesterase
    • Meng, L., Sin, N. & Crews, C. M. The antiproliferative agent didemnin B uncompetitively inhibits palmitoyl protein thioesterase. Biochemistry 37, 10488-10492 (1998).
    • (1998) Biochemistry , vol.37 , pp. 10488-10492
    • Meng, L.1    Sin, N.2    Crews, C.M.3
  • 67
    • 0029913829 scopus 로고    scopus 로고
    • Didemnin binds to the protein palmitoyl thioesterase responsible for infantile neuronal ceroid lipofuscinosis
    • Crews, C. M., Lane, W. S. & Schreiber, S. L. Didemnin binds to the protein palmitoyl thioesterase responsible for infantile neuronal ceroid lipofuscinosis. Proc. Natl Acad. Sci. USA 93, 4316-4319 (1996).
    • (1996) Proc. Natl Acad. Sci. USA , vol.93 , pp. 4316-4319
    • Crews, C.M.1    Lane, W.S.2    Schreiber, S.L.3
  • 68
    • 0021217559 scopus 로고
    • Mechanism of action of didemnin B, a depsipeptide from the sea
    • Li, L. H. et al. Mechanism of action of didemnin B, a depsipeptide from the sea. Cancer Lett. 23, 279-288 (1984).
    • (1984) Cancer Lett , vol.23 , pp. 279-288
    • Li, L.H.1
  • 69
    • 0029114876 scopus 로고
    • Mechanism of protein synthesis inhibition by didemnin B in vitro
    • SirDeshpande, B. V. & Toogood, P. L. Mechanism of protein synthesis inhibition by didemnin B in vitro. Biochemistry 34, 9177-9184 (1995).
    • (1995) Biochemistry , vol.34 , pp. 9177-9184
    • SirDeshpande, B.V.1    Toogood, P.L.2
  • 70
    • 0034681943 scopus 로고    scopus 로고
    • Inhibition of protein synthesis by didemnin B: How EF-1a mediates inhibition of translocation
    • Ahuja, D. et al. Inhibition of protein synthesis by didemnin B: how EF-1a mediates inhibition of translocation. Biochemistry 39 4339-4346 (2000).
    • (2000) Biochemistry , vol.39 , pp. 4339-4346
    • Ahuja, D.1
  • 71
    • 4143051231 scopus 로고    scopus 로고
    • Structural basis for the binding of didemnins to human elongation factor eEF1A and rationale for the potent antitumor activity of these marine natural products
    • Marco, E., Martin-Santamaria, S., Cuevas, C. & Gago, F. Structural basis for the binding of didemnins to human elongation factor eEF1A and rationale for the potent antitumor activity of these marine natural products. J. Med. Chem. 47, 4439-4452 (2004).
    • (2004) J. Med. Chem , vol.47 , pp. 4439-4452
    • Marco, E.1    Martin-Santamaria, S.2    Cuevas, C.3    Gago, F.4
  • 72
    • 0028300782 scopus 로고
    • GTP-dependent binding of the antiproliferative agent didemnin to elongation factor 1 α
    • Crews, C. M., Collins, J. L., Lane, W. S., Snapper, M. L. & Schreiber, S. L. GTP-dependent binding of the antiproliferative agent didemnin to elongation factor 1 α. J. Biol. Chem. 269, 15411-15414 (1994).
    • (1994) J. Biol. Chem , vol.269 , pp. 15411-15414
    • Crews, C.M.1    Collins, J.L.2    Lane, W.S.3    Snapper, M.L.4    Schreiber, S.L.5
  • 73
    • 0032788432 scopus 로고    scopus 로고
    • Inhibition of protein synthesis by didemnin B is not sufficient to induce apoptosis in human mammary carcinoma (MCF7) cells
    • Beidler, D. R., Ahuja, D., Wicha, M. S. & Toogood, P. L. Inhibition of protein synthesis by didemnin B is not sufficient to induce apoptosis in human mammary carcinoma (MCF7) cells. Biochem. Pharmacol. 58 1067-1074 (1999).
    • (1999) Biochem. Pharmacol , vol.58 , pp. 1067-1074
    • Beidler, D.R.1    Ahuja, D.2    Wicha, M.S.3    Toogood, P.L.4
  • 74
    • 18444411949 scopus 로고    scopus 로고
    • Cell cycle phase perturbations and apoptosis in tumour cells induced by aplidine
    • Erba, E. et al. Cell cycle phase perturbations and apoptosis in tumour cells induced by aplidine. Br. J. Cancer 86, 1510-1517 (2002).
    • (2002) Br. J. Cancer , vol.86 , pp. 1510-1517
    • Erba, E.1
  • 75
    • 0029923405 scopus 로고    scopus 로고
    • Protein tyrosine kinase inhibitors prevent didemnin B-induced apoptosis in HL-60 cells
    • Johnson, K. L., Vaillant, F. & Lawen, A. Protein tyrosine kinase inhibitors prevent didemnin B-induced apoptosis in HL-60 cells. FEBS Lett. 383, 1-5 (1996).
    • (1996) FEBS Lett , vol.383 , pp. 1-5
    • Johnson, K.L.1    Vaillant, F.2    Lawen, A.3
  • 76
    • 0033011588 scopus 로고    scopus 로고
    • Rapamycin inhibits didemnin B-induced apoptosis in human HL-60 cells: Evidence for the possible involvement of FK506-binding protein 25
    • Johnson, K. L. & Lawen, A. Rapamycin inhibits didemnin B-induced apoptosis in human HL-60 cells: Evidence for the possible involvement of FK506-binding protein 25. Immunol. Cell Biol. 77, 242-248 (1999).
    • (1999) Immunol. Cell Biol , vol.77 , pp. 242-248
    • Johnson, K.L.1    Lawen, A.2
  • 77
    • 0021079226 scopus 로고
    • Didemnins A and B. Effectiveness against cutaneous herpes simplex virus in mice
    • Weed, S. D. & Stringfellow, D. A. Didemnins A and B. Effectiveness against cutaneous herpes simplex virus in mice. Antiviral Res. 3, 269-274 (1983).
    • (1983) Antiviral Res , vol.3 , pp. 269-274
    • Weed, S.D.1    Stringfellow, D.A.2
  • 78
    • 0029670281 scopus 로고    scopus 로고
    • Antiproliferative effect of dehydrodidemnin B (DDB), a depsipeptide isolated from Mediterranean tunicates
    • Urdiales, J. L., Morata, P., Nunez De Castro, I. & Sanchez-Jimenez, F. Antiproliferative effect of dehydrodidemnin B (DDB), a depsipeptide isolated from Mediterranean tunicates. Cancer Lett. 102, 31-37 (1996).
    • (1996) Cancer Lett , vol.102 , pp. 31-37
    • Urdiales, J.L.1    Morata, P.2    Nunez De Castro, I.3    Sanchez-Jimenez, F.4
  • 79
    • 0023745136 scopus 로고
    • Phase I clinical and pharmacokinetic investigation of didemnin B, a cyclic depsipeptide
    • Dorr, F. A., Kuhn, J. G., Phillips, J. & von Hoff, D. D. Phase I clinical and pharmacokinetic investigation of didemnin B, a cyclic depsipeptide. Eur. J. Cancer Clin. Oncol. 24, 1699-1706 (1988).
    • (1988) Eur. J. Cancer Clin. Oncol , vol.24 , pp. 1699-1706
    • Dorr, F.A.1    Kuhn, J.G.2    Phillips, J.3    von Hoff, D.D.4
  • 80
    • 0023851572 scopus 로고
    • Application of a new preclinical drug screening system for cancer of the large bowel
    • Scheithauer, W., Moyer, M. P., Clark, G. M. & Von Hoff, D. D. Application of a new preclinical drug screening system for cancer of the large bowel. Cancer Chemother. Pharmacol. 21, 31-34 (1988).
    • (1988) Cancer Chemother. Pharmacol , vol.21 , pp. 31-34
    • Scheithauer, W.1    Moyer, M.P.2    Clark, G.M.3    Von Hoff, D.D.4
  • 81
    • 0028815543 scopus 로고
    • Didemnin B induces cell death by apoptosis: The fastest induction of apoptosis ever described
    • Grubb, D. R., Wolvetang, E. J. & Lawen, A. Didemnin B induces cell death by apoptosis: The fastest induction of apoptosis ever described. Biochem. Biophys. Res. Commun. 215, 1130-1136 (1995).
    • (1995) Biochem. Biophys. Res. Commun , vol.215 , pp. 1130-1136
    • Grubb, D.R.1    Wolvetang, E.J.2    Lawen, A.3
  • 82
    • 0032780130 scopus 로고    scopus 로고
    • Cytotoxicity and neurocytotoxicity of new marine anticancer agents evaluated using in vitro assays
    • Geldof, A. A., Mastbergen, S. C., Henrar, R. E. & Faircloth, G. T. Cytotoxicity and neurocytotoxicity of new marine anticancer agents evaluated using in vitro assays. Cancer Chemother. Pharmacol. 44, 312-318 (1999).
    • (1999) Cancer Chemother. Pharmacol , vol.44 , pp. 312-318
    • Geldof, A.A.1    Mastbergen, S.C.2    Henrar, R.E.3    Faircloth, G.T.4
  • 83
    • 0025748116 scopus 로고
    • A phase I clinical trial of didemnin B
    • Stewart, J. A., Low, J. B., Roberts, J. D. & Blow, A. A phase I clinical trial of didemnin B. Cancer 68, 2550-2554 (1991).
    • (1991) Cancer , vol.68 , pp. 2550-2554
    • Stewart, J.A.1    Low, J.B.2    Roberts, J.D.3    Blow, A.4
  • 85
    • 0025932957 scopus 로고
    • Phase I/II clinical trial of didemnin B in non-small-cell lung cancer: Neuromuscular toxicity is dose-limiting
    • Shin, D. M. et al. Phase I/II clinical trial of didemnin B in non-small-cell lung cancer: Neuromuscular toxicity is dose-limiting. Cancer Chemother. Pharmacol. 29, 145-149 (1991).
    • (1991) Cancer Chemother. Pharmacol , vol.29 , pp. 145-149
    • Shin, D.M.1
  • 86
    • 0026650761 scopus 로고
    • Phase II clinical and pharmacological study of didemnin B in patients with metastatic breast cancer
    • Benvenuto, J. A. et al. Phase II clinical and pharmacological study of didemnin B in patients with metastatic breast cancer. Invest. New Drugs 10, 113-117 (1992).
    • (1992) Invest. New Drugs , vol.10 , pp. 113-117
    • Benvenuto, J.A.1
  • 87
    • 0028073742 scopus 로고
    • Phase II clinical trial of didemnin B in previously treated small cell lung cancer
    • Shin, D. M. et al. Phase II clinical trial of didemnin B in previously treated small cell lung cancer. Invest. New Drugs 12 243-249 (1994).
    • (1994) Invest. New Drugs , vol.12 , pp. 243-249
    • Shin, D.M.1
  • 88
    • 0029552297 scopus 로고
    • Didemnin B in favorable histology non-Hodgkin's lymphoma: A phase II study of the National Cancer Institute of Canada Clinical Trials Group
    • Goss, G. et al. Didemnin B in favorable histology non-Hodgkin's lymphoma: A phase II study of the National Cancer Institute of Canada Clinical Trials Group. Invest. New Drugs 13, 257-260 (1995).
    • (1995) Invest. New Drugs , vol.13 , pp. 257-260
    • Goss, G.1
  • 89
    • 0034445547 scopus 로고    scopus 로고
    • Phase II trail of didemnin B in previously treated non-Hodgkin's lymphoma: An Eastern Cooperative Oncology Group (ECOG) Study
    • Kucuk, O. et al. Phase II trail 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).
    • (2000) Am. J. Clin. Oncol , vol.23 , pp. 273-277
    • Kucuk, O.1
  • 90
    • 0032413540 scopus 로고    scopus 로고
    • A phase II study of Didemnin B (NSC 325319) in advanced malignant melanoma: An Eastern Cooperative Oncology Group study (PB687)
    • Hochster, H., Oratz, R., Ettinger, D. S. & Borden, E. A phase II study of Didemnin B (NSC 325319) in advanced malignant melanoma: An Eastern Cooperative Oncology Group study (PB687). Invest. New Drugs 16 259-263 (1999).
    • (1999) Invest. New Drugs , vol.16 , pp. 259-263
    • Hochster, H.1    Oratz, R.2    Ettinger, D.S.3    Borden, E.4
  • 91
    • 0032762601 scopus 로고    scopus 로고
    • Phase II clinical trial of didemnin B in patients with recurrent or refractory anaplastic astrocytoma or glioblastoma multiforme (NSC 325319)
    • Mittelman, A. et al. Phase II clinical trial of didemnin B in patients with recurrent or refractory anaplastic astrocytoma or glioblastoma multiforme (NSC 325319). Invest. New Drugs 17, 179-182 (1999).
    • (1999) Invest. New Drugs , vol.17 , pp. 179-182
    • Mittelman, A.1
  • 92
    • 0032457858 scopus 로고    scopus 로고
    • Phase II study of didemnin B in central nervous system tumors: A Southwest Oncology Group study
    • Taylor, S. A. et al. Phase II study of didemnin B in central nervous system tumors: A Southwest Oncology Group study. Invest. New Drugs 16, 331-332 (1999).
    • (1999) Invest. New Drugs , vol.16 , pp. 331-332
    • Taylor, S.A.1
  • 93
    • 0033635276 scopus 로고    scopus 로고
    • Pharmaceutical development of anticancer agents from marine sources
    • Nuijen, B. et al. Pharmaceutical development of anticancer agents from marine sources. Anticancer Drugs 11, 793-811 (2000).
    • (2000) Anticancer Drugs , vol.11 , pp. 793-811
    • Nuijen, B.1
  • 94
    • 58149177611 scopus 로고    scopus 로고
    • Rinehart, K. L. & Lithgow-Bertelloni, A. M. Dehydrodidemnin B. WO9104985 (A1) (1991).
    • Rinehart, K. L. & Lithgow-Bertelloni, A. M. Dehydrodidemnin B. WO9104985 (A1) (1991).
  • 95
    • 3042781436 scopus 로고    scopus 로고
    • Aplidin induces the mitochondrial apoptotic pathway via oxidative stress-mediated JNK and p38 activation and protein kinase C
    • Garcia-Fernandez, L. F. et al. Aplidin induces the mitochondrial apoptotic pathway via oxidative stress-mediated JNK and p38 activation and protein kinase C. Oncogene 21, 7533-7544 (2002).
    • (2002) Oncogene , vol.21 , pp. 7533-7544
    • Garcia-Fernandez, L.F.1
  • 96
    • 0037388458 scopus 로고    scopus 로고
    • Rapid and selective apoptosis in human leukemic cells induced by Aplidine through a Fas/CD95- and mitochondrial-mediated mechanism
    • Gajate, C., An, F. & Mollinedo, F. Rapid and selective apoptosis in human leukemic cells induced by Aplidine through a Fas/CD95- and mitochondrial-mediated mechanism. Clin. Cancer Res. 9, 1535-1545 (2003).
    • (2003) Clin. Cancer Res , vol.9 , pp. 1535-1545
    • Gajate, C.1    An, F.2    Mollinedo, F.3
  • 97
    • 0035811588 scopus 로고    scopus 로고
    • Mitochondrial cytochrome c release is caspase-dependent and does not involve mitochondrial permeability transition in didemnin B-induced apoptosis
    • Grubb, D. R., Ly, J. D., Vaillant, F., Johnson, K. L. & Lawen, A. Mitochondrial cytochrome c release is caspase-dependent and does not involve mitochondrial permeability transition in didemnin B-induced apoptosis. Oncogene 20, 4085-4094 (2001).
    • (2001) Oncogene , vol.20 , pp. 4085-4094
    • Grubb, D.R.1    Ly, J.D.2    Vaillant, F.3    Johnson, K.L.4    Lawen, A.5
  • 98
    • 0346036093 scopus 로고    scopus 로고
    • Aplidin induces apoptosis in human cancer cells via glutathione depletion and sustained activation of the epidermal growth factor receptor, Src, JNK, and p38 MAPK
    • Cuadrado, A. et al. Aplidin induces apoptosis in human cancer cells via glutathione depletion and sustained activation of the epidermal growth factor receptor, Src, JNK, and p38 MAPK. J. Biol. Chem. 278, 241-250 (2003).
    • (2003) J. Biol. Chem , vol.278 , pp. 241-250
    • Cuadrado, A.1
  • 99
    • 2942724508 scopus 로고    scopus 로고
    • JNK activation is critical for Aplidin-induced apoptosis
    • Cuadrado, A., Gonzalez, L., Suarez, Y., Martinez, T. & Munoz, A. JNK activation is critical for Aplidin-induced apoptosis. Oncogene 23, 4673-4680 (2004).
    • (2004) Oncogene , vol.23 , pp. 4673-4680
    • Cuadrado, A.1    Gonzalez, L.2    Suarez, Y.3    Martinez, T.4    Munoz, A.5
  • 100
    • 0037265805 scopus 로고    scopus 로고
    • Aplidine, a new anticancer agent of marine origin, inhibits vascular endothelial growth factor (VEGF) secretion and blocks VEGF-VEGFR-1 (flt-1) autocrine loop in human leukemia cells MOLT-4
    • Broggini, M. et al. Aplidine, a new anticancer agent of marine origin, inhibits vascular endothelial growth factor (VEGF) secretion and blocks VEGF-VEGFR-1 (flt-1) autocrine loop in human leukemia cells MOLT-4. Leukemia 17, 52-59 (2003).
    • (2003) Leukemia , vol.17 , pp. 52-59
    • Broggini, M.1
  • 101
    • 27144487474 scopus 로고    scopus 로고
    • VEGF inhibition and cytotoxic effect of aplidin in leukemia cell lines and cells from acute myeloid leukemia
    • Biscardi, M. et al. VEGF inhibition and cytotoxic effect of aplidin in leukemia cell lines and cells from acute myeloid leukemia. Ann. Oncol. 16, 1667-1674 (2005).
    • (2005) Ann. Oncol , vol.16 , pp. 1667-1674
    • Biscardi, M.1
  • 102
    • 6944232399 scopus 로고    scopus 로고
    • Establishment and characterisation of a human carcinoma cell line with acquired resistance to Aplidin
    • Losada, A., Lopez-Oliva, J. M., Sanchez-Puelles, J. M. & Garcia-Fernandez, L. F. Establishment and characterisation of a human carcinoma cell line with acquired resistance to Aplidin. Br. J. Cancer 91, 1405-1413 (2004).
    • (2004) Br. J. Cancer , vol.91 , pp. 1405-1413
    • Losada, A.1    Lopez-Oliva, J.M.2    Sanchez-Puelles, J.M.3    Garcia-Fernandez, L.F.4
  • 103
    • 0041733458 scopus 로고    scopus 로고
    • Effect of aplidine in acute lymphoblastic leukaemia cells
    • Erba, E. et al. Effect of aplidine in acute lymphoblastic leukaemia cells. Br. J. Cancer 89, 763-773 (2003).
    • (2003) Br. J. Cancer , vol.89 , pp. 763-773
    • Erba, E.1
  • 104
    • 0036288919 scopus 로고    scopus 로고
    • In vitro toxicity of ET-743 and aplidine, two marine-derived antineoplastics, on human bone marrow haematopoietic progenitors. comparison with the clinical results
    • Albella, B. et al. In vitro toxicity of ET-743 and aplidine, two marine-derived antineoplastics, on human bone marrow haematopoietic progenitors. comparison with the clinical results. Eur. J. Cancer 38, 1395-1404 (2002).
    • (2002) Eur. J. Cancer , vol.38 , pp. 1395-1404
    • Albella, B.1
  • 105
    • 0036790679 scopus 로고    scopus 로고
    • Didemnin B induces apoptosis in proliferating but not resting peripheral blood mononuclear cells
    • Baker, M. A., Grubb, D. R. & Lawen, A. Didemnin B induces apoptosis in proliferating but not resting peripheral blood mononuclear cells. Apoptosis 7, 407-412 (2002).
    • (2002) Apoptosis , vol.7 , pp. 407-412
    • Baker, M.A.1    Grubb, D.R.2    Lawen, A.3
  • 106
    • 0037766254 scopus 로고    scopus 로고
    • In vitro cytotoxicity of aplidin and crossresistance with other cytotoxic drugs in childhood leukemic and normal bone marrow and blood samples: A rational basis for clinical development
    • Bresters, D. et al. In vitro cytotoxicity of aplidin and crossresistance with other cytotoxic drugs in childhood leukemic and normal bone marrow and blood samples: A rational basis for clinical development. Leukemia 17, 1338-1343 (2003).
    • (2003) Leukemia , vol.17 , pp. 1338-1343
    • Bresters, D.1
  • 107
    • 0142227045 scopus 로고    scopus 로고
    • In vitro toxicity of three new antitumoral drugs (trabectedin, aplidin, and kahalalide F) on hematopoietic progenitors and stem cells
    • Gomez, S. G., Bueren, J. A., Faircloth, G. T., Jimeno, J. & Albella, B. In vitro toxicity of three new antitumoral drugs (trabectedin, aplidin, and kahalalide F) on hematopoietic progenitors and stem cells. Exp. Hematol. 31, 1104-1111 (2003).
    • (2003) Exp. Hematol , vol.31 , pp. 1104-1111
    • Gomez, S.G.1    Bueren, J.A.2    Faircloth, G.T.3    Jimeno, J.4    Albella, B.5
  • 108
    • 3042743711 scopus 로고    scopus 로고
    • Antiangiogenic activity of aplidine, a new agent of marine origin
    • Taraboletti, G. et al. Antiangiogenic activity of aplidine, a new agent of marine origin. Br. J. Cancer 90, 2418-2424 (2004).
    • (2004) Br. J. Cancer , vol.90 , pp. 2418-2424
    • Taraboletti, G.1
  • 109
    • 0642345073 scopus 로고    scopus 로고
    • Inhibition by Aplidine of the aggregation of the prion peptide PrP 106-126 into β-sheet fibrils
    • Perez, M., Sadqi, M., Munoz, V. & Avila, J. Inhibition by Aplidine of the aggregation of the prion peptide PrP 106-126 into β-sheet fibrils. Biochem. Biophys. Acta 1639, 133-139 (2003).
    • (2003) Biochem. Biophys. Acta , vol.1639 , pp. 133-139
    • Perez, M.1    Sadqi, M.2    Munoz, V.3    Avila, J.4
  • 110
    • 0028305511 scopus 로고
    • Hemiasterlin and geodiamolide TA: Two new cytotoxic peptides from the marine sponge Hemiasterella minor (Kirkpatrick)
    • Talpir, R., Benayahu, Y., Kashman, Y., Pannell, L. & Schleyer, M. Hemiasterlin and geodiamolide TA: Two new cytotoxic peptides from the marine sponge Hemiasterella minor (Kirkpatrick). Tetrahedron Lett. 35, 4453-4456 (1994).
    • (1994) Tetrahedron Lett , vol.35 , pp. 4453-4456
    • Talpir, R.1    Benayahu, Y.2    Kashman, Y.3    Pannell, L.4    Schleyer, M.5
  • 113
    • 0032769659 scopus 로고    scopus 로고
    • Cytotoxic and tubulin-interactive hemiasterlins from Auletta sp. and Siphonochalina spp. sponges
    • Gamble, W. R. et al. Cytotoxic and tubulin-interactive hemiasterlins from Auletta sp. and Siphonochalina spp. sponges. Bioorg. Med. Chem. 7, 1611-1615 (1999).
    • (1999) Bioorg. Med. Chem , vol.7 , pp. 1611-1615
    • Gamble, W.R.1
  • 114
    • 0031060389 scopus 로고    scopus 로고
    • Cytotoxic peptides hemiasterlin, hemiasterlin A and hemiasterlin B induce mitotic arrest and abnormal spindle formation
    • Anderson, H. J., Coleman, J. E., Andersen, R. J. & Roberge, M. Cytotoxic peptides hemiasterlin, hemiasterlin A and hemiasterlin B induce mitotic arrest and abnormal spindle formation. Cancer Chemother. Pharmacol. 39, 223-226 (1997).
    • (1997) Cancer Chemother. Pharmacol , vol.39 , pp. 223-226
    • Anderson, H.J.1    Coleman, J.E.2    Andersen, R.J.3    Roberge, M.4
  • 115
    • 0033607202 scopus 로고    scopus 로고
    • Interactions of the sponge-derived antimitotic tripeptide hemiasterlin with tubulin: Comparison with dolastatin 10 and cryptophycin 1
    • Bai, R., Durso, N. A., Sackett, D. L. & Hamel, E. Interactions of the sponge-derived antimitotic tripeptide hemiasterlin with tubulin: comparison with dolastatin 10 and cryptophycin 1. Biochemistry 38, 14302-14310 (1999).
    • (1999) Biochemistry , vol.38 , pp. 14302-14310
    • Bai, R.1    Durso, N.A.2    Sackett, D.L.3    Hamel, E.4
  • 116
    • 0037331338 scopus 로고    scopus 로고
    • Synthesis and antimitotic/cytotoxic activity of hemiasterlin analogues
    • Nieman, J. A. et al. Synthesis and antimitotic/cytotoxic activity of hemiasterlin analogues. J. Nat. Prod. 66, 183-199 (2003).
    • (2003) J. Nat. Prod , vol.66 , pp. 183-199
    • Nieman, J.A.1
  • 117
    • 4444352764 scopus 로고    scopus 로고
    • 1,3-dimethyl-L-valinamide (HTI-286)
    • 1,3-dimethyl-L-valinamide (HTI-286). J. Med. Chem. 47, 4774-4786 (2004).
    • (2004) J. Med. Chem , vol.47 , pp. 4774-4786
    • Zask, A.1
  • 118
    • 3142661760 scopus 로고    scopus 로고
    • D-piece modifications of the hemiasterlin analog HTI-286 produce potent tubulin inhibitors
    • Zask, A. et al. D-piece modifications of the hemiasterlin analog HTI-286 produce potent tubulin inhibitors. Bioorg. Med. Chem. Lett. 14, 4353-4358 (2004).
    • (2004) Bioorg. Med. Chem. Lett , vol.14 , pp. 4353-4358
    • Zask, A.1
  • 119
    • 5344252376 scopus 로고    scopus 로고
    • Synthesis and activity of novel analogs of hemiasterlin as inhibitors of tubulin polymerization: Modification of the A segment
    • Yamashita, A. et al. Synthesis and activity of novel analogs of hemiasterlin as inhibitors of tubulin polymerization: Modification of the A segment. Bioorg. Med. Chem. Lett. 14, 5317-5322 (2004).
    • (2004) Bioorg. Med. Chem. Lett , vol.14 , pp. 5317-5322
    • Yamashita, A.1
  • 120
    • 3142755555 scopus 로고    scopus 로고
    • Tubulin inhibitors. Synthesis and biological activity of HTI-286 analogs with B-segment heterosubstituents
    • Niu, C. et al. Tubulin inhibitors. Synthesis and biological activity of HTI-286 analogs with B-segment heterosubstituents. Bioorg. Med. Chem. Lett. 14, 4329-4332 (2004).
    • (2004) Bioorg. Med. Chem. Lett , vol.14 , pp. 4329-4332
    • Niu, C.1
  • 121
    • 4043138633 scopus 로고    scopus 로고
    • Probing the interaction of HTI-286 with tubulin using a stilbene analogue
    • Lo, M. C. et al. Probing the interaction of HTI-286 with tubulin using a stilbene analogue. J. Am. Chem. Soc. 126, 9898-9899 (2004).
    • (2004) J. Am. Chem. Soc , vol.126 , pp. 9898-9899
    • Lo, M.C.1
  • 122
    • 0345254942 scopus 로고    scopus 로고
    • Biophysical characterization of the interactions of HTI-286 with tubulin heterodimer and microtubules
    • Krishnamurthy, G. et al. Biophysical characterization of the interactions of HTI-286 with tubulin heterodimer and microtubules. Biochemistry 42, 13484-13495 (2003).
    • (2003) Biochemistry , vol.42 , pp. 13484-13495
    • Krishnamurthy, G.1
  • 123
    • 8344289232 scopus 로고    scopus 로고
    • Localization of the antimitotic peptide and depsipeptide binding site on β-tubulin
    • Mitra, A. & Sept, D. Localization of the antimitotic peptide and depsipeptide binding site on β-tubulin. Biochemistry 43, 13955-13962 (2004).
    • (2004) Biochemistry , vol.43 , pp. 13955-13962
    • Mitra, A.1    Sept, D.2
  • 124
    • 20944436156 scopus 로고    scopus 로고
    • Two photoaffinity analogues of the tripeptide, hemiasterlin, exclusively label α-tubulin
    • Nunes, M. et al. Two photoaffinity analogues of the tripeptide, hemiasterlin, exclusively label α-tubulin. Biochemistry 44, 6844-6857 (2005).
    • (2005) Biochemistry , vol.44 , pp. 6844-6857
    • Nunes, M.1
  • 125
    • 7444224866 scopus 로고    scopus 로고
    • Tumor cells resistant to a microtubule-depolymerizing hemiasterlin analogue, HTI-286, have mutations in α- or β-tubulin and increased microtubule stability
    • Poruchynsky, M. S. et al. Tumor cells resistant to a microtubule-depolymerizing hemiasterlin analogue, HTI-286, have mutations in α- or β-tubulin and increased microtubule stability. Biochemistry 43, 13944-13954 (2004).
    • (2004) Biochemistry , vol.43 , pp. 13944-13954
    • Poruchynsky, M.S.1
  • 126
    • 7444235448 scopus 로고    scopus 로고
    • Cells resistant to HTI-286 do not overexpress P-glycoprotein but have reduced drug accumulation and a point mutation in α-tubulin
    • Loganzo, F. et al. Cells resistant to HTI-286 do not overexpress P-glycoprotein but have reduced drug accumulation and a point mutation in α-tubulin. Mol. Cancer Ther. 3, 1319-1327 (2004).
    • (2004) Mol. Cancer Ther , vol.3 , pp. 1319-1327
    • Loganzo, F.1
  • 127
    • 0037447174 scopus 로고    scopus 로고
    • HTI-286, a synthetic analogue of the tripeptide hemiasterlin, is a potent antimicrotubule agent that circumvents P-glycoprotein-mediated resistance in vitro and in vivo
    • Loganzo, F. et al. HTI-286, a synthetic analogue of the tripeptide hemiasterlin, is a potent antimicrotubule agent that circumvents P-glycoprotein-mediated resistance in vitro and in vivo. Cancer Res. 63, 1838-1845 (2003).
    • (2003) Cancer Res , vol.63 , pp. 1838-1845
    • Loganzo, F.1
  • 128
    • 3142657011 scopus 로고    scopus 로고
    • Phase 1 and pharmacological study of HTI-286, a novel antimicrotubule agent: Correlation of neutropenia with time above a threshold serum concentration
    • Ratain M. J. et al. 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).
    • (2003) Proc. Am. Soc. Clin. Oncol , vol.22 , pp. 516
    • Ratain, M.J.1
  • 129
    • 42149107394 scopus 로고    scopus 로고
    • Targeting prostate cancer with HTI-286, a synthetic analog of the marine sponge product hemiasterlin
    • Hadaschik, B. A. et al. Targeting prostate cancer with HTI-286, a synthetic analog of the marine sponge product hemiasterlin Int. J. Cancer: 122, 2368-2376 (2008).
    • (2008) Int. J. Cancer , vol.122 , pp. 2368-2376
    • Hadaschik, B.A.1
  • 130
    • 0034597447 scopus 로고    scopus 로고
    • The chemistry and biology of discodermolide
    • Kalesse, M. The chemistry and biology of discodermolide. ChemBiochem 1, 171-175 (2000).
    • (2000) ChemBiochem , vol.1 , pp. 171-175
    • Kalesse, M.1
  • 131
    • 4544303502 scopus 로고    scopus 로고
    • (+)-discodermolide: A marine natural product against cancer
    • De Souza, M. V. (+)-discodermolide: A marine natural product against cancer. Scientific World Journal 4, 415-436 (2004).
    • (2004) Scientific World Journal , vol.4 , pp. 415-436
    • De Souza, M.V.1
  • 132
    • 0025942924 scopus 로고
    • Discodermolide - a new, marine-derived immunosuppressive compound. I. In vitro studies
    • Longley, R. E., Caddigan, D., Harmody, D., Gunasekera, M. & Gunasekera, S. P. Discodermolide - a new, marine-derived immunosuppressive compound. I. In vitro studies. Transplantation 52, 650-656 (1991).
    • (1991) Transplantation , vol.52 , pp. 650-656
    • Longley, R.E.1    Caddigan, D.2    Harmody, D.3    Gunasekera, M.4    Gunasekera, S.P.5
  • 133
    • 0025936629 scopus 로고
    • Discodermolide - a new, marine-derived immunosuppressive compound. II. In vivo studies
    • Longley, R. E., Caddigan, D., Harmody, D., Gunasekera, M. & Gunasekera, S. P. Discodermolide - a new, marine-derived immunosuppressive compound. II. In vivo studies. Transplantation 52, 656-661 (1991).
    • (1991) Transplantation , vol.52 , pp. 656-661
    • Longley, R.E.1    Caddigan, D.2    Harmody, D.3    Gunasekera, M.4    Gunasekera, S.P.5
  • 135
    • 0025160288 scopus 로고
    • Discodermolide: A new bioactive polyhydroxylated lactone from the marine sponge Discodermia dissoluta
    • Gunasekera, S. P., Gunasekera, M., Longley, R. E. & Schulte, G. K. Discodermolide: A new bioactive polyhydroxylated lactone from the marine sponge Discodermia dissoluta. J. Org. Chem. 55, 4912-4915 (1990).
    • (1990) J. Org. Chem , vol.55 , pp. 4912-4915
    • Gunasekera, S.P.1    Gunasekera, M.2    Longley, R.E.3    Schulte, G.K.4
  • 136
    • 85022401932 scopus 로고    scopus 로고
    • Gunasekera, S. P., Gunasekera, M., Longley, R. E. & Schulte, G. K. Discodermolide: A new bioactive polyhydroxylated lactone from the marine sponge Discodermia dissoluta [Erratum to document cited in CA113(9):75187b]. J. Org. Chem. 56, 1346 (1991).
    • Gunasekera, S. P., Gunasekera, M., Longley, R. E. & Schulte, G. K. Discodermolide: A new bioactive polyhydroxylated lactone from the marine sponge Discodermia dissoluta [Erratum to document cited in CA113(9):75187b]. J. Org. Chem. 56, 1346 (1991).
  • 137
    • 0027858856 scopus 로고
    • Total synthesis of the immunosuppressive agent (-)-discodermolide
    • Nerenberg, J. B., Hung, D. T., Somers, P. K. & Schreiber, S. L. Total synthesis of the immunosuppressive agent (-)-discodermolide. J. Am. Chem. Soc. 115, 12621-12622 (1993).
    • (1993) J. Am. Chem. Soc , vol.115 , pp. 12621-12622
    • Nerenberg, J.B.1    Hung, D.T.2    Somers, P.K.3    Schreiber, S.L.4
  • 138
    • 0028505014 scopus 로고
    • Distinct binding and cellular properties of synthetic (+)- and (-)-discodermolides
    • Hung, D. T., Nerenberg, J. B. & Schreiber, S. L. Distinct binding and cellular properties of synthetic (+)- and (-)-discodermolides. Chem. Biol. 1, 67-71 (1994).
    • (1994) Chem. Biol , vol.1 , pp. 67-71
    • Hung, D.T.1    Nerenberg, J.B.2    Schreiber, S.L.3
  • 139
    • 0042510054 scopus 로고    scopus 로고
    • Total synthesis of the potent microtubule-stabilizing agent (+)-discodermolide
    • Harried, S. S., Lee, C. P., Yang, G., Lee, T. I. & Myles, D. C. Total synthesis of the potent microtubule-stabilizing agent (+)-discodermolide. J. Org. Chem. 68, 6646-6660 (2003).
    • (2003) J. Org. Chem , vol.68 , pp. 6646-6660
    • Harried, S.S.1    Lee, C.P.2    Yang, G.3    Lee, T.I.4    Myles, D.C.5
  • 140
    • 11844266790 scopus 로고    scopus 로고
    • Total synthesis of (+)-discodermolide: An improved endgame exploiting a Still-Gennari-type olefination with a C1-C8 β-ketophosphonate fragment
    • Paterson, I. & Lyothier, I. Total synthesis of (+)-discodermolide: An improved endgame exploiting a Still-Gennari-type olefination with a C1-C8 β-ketophosphonate fragment. Org. Lett. 6, 4933-4936 (2004).
    • (2004) Org. Lett , vol.6 , pp. 4933-4936
    • Paterson, I.1    Lyothier, I.2
  • 141
    • 11844282737 scopus 로고    scopus 로고
    • A second-generation total synthesis of (+)-discodermolide: The development of a practical route using solely substrate-based stereocontrol
    • Paterson, I. et al. A second-generation total synthesis of (+)-discodermolide: The development of a practical route using solely substrate-based stereocontrol. J. Org. Chem. 70, 150-160 (2005).
    • (2005) J. Org. Chem , vol.70 , pp. 150-160
    • Paterson, I.1
  • 143
    • 0034644383 scopus 로고    scopus 로고
    • Evolution of a gram-scale synthesis of (+)-discodermolide
    • Smith, A. B. 3rd et al. Evolution of a gram-scale synthesis of (+)-discodermolide. J. Am. Chem. Soc. 122, 8654-8664 (2000).
    • (2000) J. Am. Chem. Soc , vol.122 , pp. 8654-8664
    • Smith 3rd, A.B.1
  • 144
    • 0344065680 scopus 로고    scopus 로고
    • A practical improvement, enhancing the large-scale synthesis of (+)-discodermolide: A third-generation approach
    • Smith, A. B. 3rd, Freeze, B. S., Brouard, I. & Hirose, T. A practical improvement, enhancing the large-scale synthesis of (+)-discodermolide: a third-generation approach. Org. Lett. 5, 4405-4408 (2003).
    • (2003) Org. Lett , vol.5 , pp. 4405-4408
    • Smith 3rd, A.B.1    Freeze, B.S.2    Brouard, I.3    Hirose, T.4
  • 145
    • 0842285267 scopus 로고    scopus 로고
    • Large-scale synthesis of the anti-cancer marine natural product (+)-discodermolide. Part 1: Synthetic strategy and preparation of a common precursor
    • Mickel, S. J. et al. Large-scale synthesis of the anti-cancer marine natural product (+)-discodermolide. Part 1: Synthetic strategy and preparation of a common precursor. Org. Proc. Res. Dev. 8 92-100 (2004).
    • (2004) Org. Proc. Res. Dev , vol.8 , pp. 92-100
    • Mickel, S.J.1
  • 146
    • 0842263632 scopus 로고    scopus 로고
    • Large-scale synthesis of the anti-cancer marine natural product (+)-discodermolide. Part 2: Synthesis of fragments C1-6 and C9-14
    • Mickel, S. J. et al. Large-scale synthesis of the anti-cancer marine natural product (+)-discodermolide. Part 2: Synthesis of fragments C1-6 and C9-14 Org. Proc. Res. Dev. 8, 101-106 (2004).
    • (2004) Org. Proc. Res. Dev , vol.8 , pp. 101-106
    • Mickel, S.J.1
  • 147
    • 0842285266 scopus 로고    scopus 로고
    • Large-scale synthesis of the anti-cancer marine natural product (+)-discodermolide. Part 3: Synthesis of fragment C15-21
    • Mickel, S. J. et al. Large-scale synthesis of the anti-cancer marine natural product (+)-discodermolide. Part 3: Synthesis of fragment C15-21 Org. Proc. Res. Dev. 8, 107-112 (2004).
    • (2004) Org. Proc. Res. Dev , vol.8 , pp. 107-112
    • Mickel, S.J.1
  • 148
    • 0842328441 scopus 로고    scopus 로고
    • Large-scale synthesis of the anti-cancer marine natural product (+)-discodermolide. Part 4: Preparation of fragment C7-24
    • Mickel, S. J. et al. Large-scale synthesis of the anti-cancer marine natural product (+)-discodermolide. Part 4: Preparation of fragment C7-24 Org. Proc. Res. Dev. 8, 113-121 (2004).
    • (2004) Org. Proc. Res. Dev , vol.8 , pp. 113-121
    • Mickel, S.J.1
  • 149
    • 0842306927 scopus 로고    scopus 로고
    • Large-scale synthesis of the anti-cancer marine natural product (+)-discodermolide. Part 5: Linkage of fragments C1-6 and C7-24 and finale
    • Mickel, S. J. et al. Large-scale synthesis of the anti-cancer marine natural product (+)-discodermolide. Part 5: Linkage of fragments C1-6 and C7-24 and finale. Org. Proc. Res. Dev. 8, 122-130 (2004).
    • (2004) Org. Proc. Res. Dev , vol.8 , pp. 122-130
    • Mickel, S.J.1
  • 150
    • 0030113284 scopus 로고    scopus 로고
    • (+)-Discodermolide binds to microtubules in stoichiometric ratio to tubulin dimers, blocks taxol binding and results in mitotic arrest
    • Hung, D. T., Chen, J. & Schreiber, S. L. (+)-Discodermolide binds to microtubules in stoichiometric ratio to tubulin dimers, blocks taxol binding and results in mitotic arrest. Chem. Biol. 3, 287-293 (1996).
    • (1996) Chem. Biol , vol.3 , pp. 287-293
    • Hung, D.T.1    Chen, J.2    Schreiber, S.L.3
  • 151
    • 0030039669 scopus 로고    scopus 로고
    • Discodermolide, a cytotoxic marine agent that stabilizes microtubules more potently than taxol
    • ter Haar, E. et al. Discodermolide, a cytotoxic marine agent that stabilizes microtubules more potently than taxol. Biochemistry 35, 243-250 (1996).
    • (1996) Biochemistry , vol.35 , pp. 243-250
    • ter Haar, E.1
  • 152
    • 0030761586 scopus 로고    scopus 로고
    • The microtubule-stabilizing agent discodermolide competitively inhibits the binding of paclitaxel (Taxol) to tubulin polymers, enhances tubulin nucleation reactions more potently than paclitaxel, and inhibits the growth of paclitaxel-resistant cells
    • Kowalski, R. J. et al. The microtubule-stabilizing agent discodermolide competitively inhibits the binding of paclitaxel (Taxol) to tubulin polymers, enhances tubulin nucleation reactions more potently than paclitaxel, and inhibits the growth of paclitaxel-resistant cells. Mol. Pharmacol. 52, 613-622 (1997).
    • (1997) Mol. Pharmacol , vol.52 , pp. 613-622
    • Kowalski, R.J.1
  • 153
    • 18844469806 scopus 로고    scopus 로고
    • Taxol and discodermolide represent a synergistic drug combination in human carcinoma cell lines
    • Martello, L. A. et al. Taxol and discodermolide represent a synergistic drug combination in human carcinoma cell lines. Clin. Cancer Res. 6, 1978-1987 (2000).
    • (2000) Clin. Cancer Res , vol.6 , pp. 1978-1987
    • Martello, L.A.1
  • 154
    • 0034071993 scopus 로고    scopus 로고
    • Discodermolide: Just another microtubule-stabilizing agent? No! A lesson in synergy
    • Giannakakou, P. & Fojo, T. Discodermolide: Just another microtubule-stabilizing agent? No! A lesson in synergy. Clin. Cancer Res. 6, 1613-1615 (2000).
    • (2000) Clin. Cancer Res , vol.6 , pp. 1613-1615
    • Giannakakou, P.1    Fojo, T.2
  • 155
    • 3142723291 scopus 로고    scopus 로고
    • Synergistic suppression of microtubule dynamics by discodermolide and paclitaxel in non-small cell lung carcinoma cells
    • Honore, S. et al. Synergistic suppression of microtubule dynamics by discodermolide and paclitaxel in non-small cell lung carcinoma cells. Cancer Res. 64, 4957-4964 (2004).
    • (2004) Cancer Res , vol.64 , pp. 4957-4964
    • Honore, S.1
  • 156
    • 0036531790 scopus 로고    scopus 로고
    • Differential mitotic responses to microtubule-stabilizing and -destabilizing drugs
    • Chen, J. G. & Horwitz, S. B. Differential mitotic responses to microtubule-stabilizing and -destabilizing drugs. Cancer Res. 62, 1935-1938 (2002).
    • (2002) Cancer Res , vol.62 , pp. 1935-1938
    • Chen, J.G.1    Horwitz, S.B.2
  • 157
    • 0037099675 scopus 로고    scopus 로고
    • Late activation of apoptotic pathways plays a negligible role in mediating the cytotoxic effects of discodermolide and epothilone B in non-small cell lung cancer cells
    • Broker, L. E. et al. Late activation of apoptotic pathways plays a negligible role in mediating the cytotoxic effects of discodermolide and epothilone B in non-small cell lung cancer cells. Cancer Res. 62, 4081-4088 (2002).
    • (2002) Cancer Res , vol.62 , pp. 4081-4088
    • Broker, L.E.1
  • 158
    • 1942490811 scopus 로고    scopus 로고
    • Suppression of microtubule dynamics by discodermolide by a novel mechanism is associated with mitotic arrest and inhibition of tumor cell proliferation
    • Honore, S. et al. Suppression of microtubule dynamics by discodermolide by a novel mechanism is associated with mitotic arrest and inhibition of tumor cell proliferation. Mol. Cancer Ther. 2 1303-1311 (2003).
    • (2003) Mol. Cancer Ther , vol.2 , pp. 1303-1311
    • Honore, S.1
  • 159
    • 0037468616 scopus 로고    scopus 로고
    • Discodermolide interferes with the binding of tau protein to microtubules
    • Kar, S., Florence, G. J., Paterson, I. & Amos, L. A. Discodermolide interferes with the binding of tau protein to microtubules. FEBS Lett. 539, 34-36 (2003).
    • (2003) FEBS Lett , vol.539 , pp. 34-36
    • Kar, S.1    Florence, G.J.2    Paterson, I.3    Amos, L.A.4
  • 160
    • 25444465116 scopus 로고    scopus 로고
    • Both microtubule-stabilizing and microtubule-destabilizing drugs inhibit hypoxia-inducible factor-1α accumulation and activity by disrupting microtubule function
    • Escuin, D., Kline, E. R. & Giannakakou, P. Both microtubule-stabilizing and microtubule-destabilizing drugs inhibit hypoxia-inducible factor-1α accumulation and activity by disrupting microtubule function. Cancer Res. 65, 9021-9028 (2005).
    • (2005) Cancer Res , vol.65 , pp. 9021-9028
    • Escuin, D.1    Kline, E.R.2    Giannakakou, P.3
  • 161
    • 21744449331 scopus 로고    scopus 로고
    • The microtubule stabilizing agent discodermolide is a potent inducer of accelerated cell senescence
    • Klein, L. E., Freeze, B. S., Smith, A. B. & Horwitz, S. B. The microtubule stabilizing agent discodermolide is a potent inducer of accelerated cell senescence. Cell Cycle 4, 501-507 (2005).
    • (2005) Cell Cycle , vol.4 , pp. 501-507
    • Klein, L.E.1    Freeze, B.S.2    Smith, A.B.3    Horwitz, S.B.4
  • 162
    • 21744455884 scopus 로고    scopus 로고
    • Discodermolide analogues as the chemical component of combination bacteriolytic therapy
    • Smith, A. B. 3rd et al. Discodermolide analogues as the chemical component of combination bacteriolytic therapy. Bioorg. Med. Chem. Lett. 15, 3623-3626 (2005).
    • (2005) Bioorg. Med. Chem. Lett , vol.15 , pp. 3623-3626
    • Smith 3rd, A.B.1
  • 163
    • 31544473618 scopus 로고    scopus 로고
    • A phase I pharmacokinetic (PK) trial of XAA296A (Discodermolide) administered every 3 wks to adult patients with advanced solid malignancies
    • Mita, A. et al. A phase I pharmacokinetic (PK) trial of XAA296A (Discodermolide) administered every 3 wks to adult patients with advanced solid malignancies. J. Clin. Oncol. 22, 2025 (2004).
    • (2004) J. Clin. Oncol , vol.22 , pp. 2025
    • Mita, A.1
  • 164
    • 0025367359 scopus 로고
    • Pharmaceuticals from cultured algae
    • Schwartz, R. E. et al. Pharmaceuticals from cultured algae. J. Ind. Microbiol. 5, 113-124 (1990).
    • (1990) J. Ind. Microbiol , vol.5 , pp. 113-124
    • Schwartz, R.E.1
  • 165
    • 0028017485 scopus 로고
    • Total structures of cryptophycins, potent antitumor depsipeptides from the blue-green-alga Nostoc sp strain Gsv-224
    • Trimurtulu, G. et al. Total structures of cryptophycins, potent antitumor depsipeptides from the blue-green-alga Nostoc sp strain Gsv-224. J. Am. Chem. Soc. 116, 4729-4737 (1994).
    • (1994) J. Am. Chem. Soc , vol.116 , pp. 4729-4737
    • Trimurtulu, G.1
  • 166
    • 0028117690 scopus 로고
    • Arenastatin A, a potent cytotoxic depsipeptide from the Okinawan marine sponge Dysidea arenaria
    • Kobayashi, M. et al. Arenastatin A, a potent cytotoxic depsipeptide from the Okinawan marine sponge Dysidea arenaria. Tetrahedron Lett. 35, 7969-7972 (1994).
    • (1994) Tetrahedron Lett , vol.35 , pp. 7969-7972
    • Kobayashi, M.1
  • 167
    • 0028962299 scopus 로고
    • Total synthesis of cryptophycins - revision of the structures of cryptophycin-A and cryptophycin-C
    • Barrow, R. A. et al. Total synthesis of cryptophycins - revision of the structures of cryptophycin-A and cryptophycin-C. J. Am. Chem. Soc. 117, 2479-2490 (1995).
    • (1995) J. Am. Chem. Soc , vol.117 , pp. 2479-2490
    • Barrow, R.A.1
  • 168
    • 0034913052 scopus 로고    scopus 로고
    • Cryptophycin-induced hyper-phosphorylation of Bcl-2, cell cycle arrest and growth inhibition in human H460 NSCLC cells
    • Lu, K., Dempsey, J., Schultz, R. M., Shih, C. & Teicher, B. A. Cryptophycin-induced hyper-phosphorylation of Bcl-2, cell cycle arrest and growth inhibition in human H460 NSCLC cells. Cancer Chemother. Pharmacol. 47, 170-178 (2001).
    • (2001) Cancer Chemother. Pharmacol , vol.47 , pp. 170-178
    • Lu, K.1    Dempsey, J.2    Schultz, R.M.3    Shih, C.4    Teicher, B.A.5
  • 169
    • 0030272586 scopus 로고    scopus 로고
    • Cryptophycin 1 binds to tubulin at a site distinct from the colchicine binding site and at a site that may overlap the vinca binding site
    • Mooberry, S. L., Taoka, C. R. & Busquets, L. Cryptophycin 1 binds to tubulin at a site distinct from the colchicine binding site and at a site that may overlap the vinca binding site. Cancer Lett. 107 53-57 (1996).
    • (1996) Cancer Lett , vol.107 , pp. 53-57
    • Mooberry, S.L.1    Taoka, C.R.2    Busquets, L.3
  • 171
    • 0029874514 scopus 로고    scopus 로고
    • Mechanism of action of cryptophycin - interaction with the vinca alkaloid domain of tubulin
    • Smith, C. D. & Zhang, X. Q. Mechanism of action of cryptophycin - interaction with the vinca alkaloid domain of tubulin. J. Biol. Chem. 271, 6192-6198 (1996).
    • (1996) J. Biol. Chem , vol.271 , pp. 6192-6198
    • Smith, C.D.1    Zhang, X.Q.2
  • 172
    • 0030721014 scopus 로고    scopus 로고
    • Mechanism of action of the unusually potent microtubule inhibitor cryptophycin 1
    • Panda, D., Himes, R. H., Moore, R. E., Wilson, L. & Jordan, M. A. Mechanism of action of the unusually potent microtubule inhibitor cryptophycin 1. Biochemistry 36, 12948-12953 (1997).
    • (1997) Biochemistry , vol.36 , pp. 12948-12953
    • Panda, D.1    Himes, R.H.2    Moore, R.E.3    Wilson, L.4    Jordan, M.A.5
  • 173
    • 0032903928 scopus 로고    scopus 로고
    • In vitro pharmacology of cryptophycin 52 (LY355703) in human tumor cell lines
    • Wagner, M. M. et al. In vitro pharmacology of cryptophycin 52 (LY355703) in human tumor cell lines. Cancer Chemother. Pharmacol. 43, 115-125 (1999).
    • (1999) Cancer Chemother. Pharmacol , vol.43 , pp. 115-125
    • Wagner, M.M.1
  • 174
    • 0036892740 scopus 로고    scopus 로고
    • Phase I and pharmacological studies of the cryptophycin analogue LY355703 administered on a single intermittent or weekly schedule
    • Sessa, C. et al. Phase I and pharmacological studies of the cryptophycin analogue LY355703 administered on a single intermittent or weekly schedule. Eur. J. Cancer 38, 2388-2396 (2002).
    • (2002) Eur. J. Cancer , vol.38 , pp. 2388-2396
    • Sessa, C.1
  • 175
    • 0036023410 scopus 로고    scopus 로고
    • Phase I trial of the cryptophycin analogue LY355703 administered as an intravenous infusion on a day 1 and 8 schedule every 21 days
    • Stevenson, J. P. et al. Phase I trial of the cryptophycin analogue LY355703 administered as an intravenous infusion on a day 1 and 8 schedule every 21 days. Clin. Cancer Res. 8, 2524-2529 (2002).
    • (2002) Clin. Cancer Res , vol.8 , pp. 2524-2529
    • Stevenson, J.P.1
  • 176
    • 0037316411 scopus 로고    scopus 로고
    • Phase 2 study of cryptophycin 52 (LY355703) in patients previously treated with platinum based chemotherapy for advanced non-small cell lung cancer
    • Edelman, M. J. et al. Phase 2 study of cryptophycin 52 (LY355703) in patients previously treated with platinum based chemotherapy for advanced non-small cell lung cancer. Lung Cancer 39, 197-199 (2003).
    • (2003) Lung Cancer , vol.39 , pp. 197-199
    • Edelman, M.J.1
  • 177
    • 21044434617 scopus 로고    scopus 로고
    • Cryptophycins-309, 249 and other cryptophycin analogs: Preclinical efficacy studies with mouse and human tumors
    • Liang, J. et al. Cryptophycins-309, 249 and other cryptophycin analogs: Preclinical efficacy studies with mouse and human tumors. Investig. New Drugs 23, 213-224 (2005).
    • (2005) Investig. New Drugs , vol.23 , pp. 213-224
    • Liang, J.1
  • 179
    • 0023584049 scopus 로고
    • The isolation and structure of a remarkable marine animal antineoplastic constituent - Dolastatin 10
    • Pettit, G. R. et al. The isolation and structure of a remarkable marine animal antineoplastic constituent - Dolastatin 10. J. Am. Chem. Soc. 109, 6883-6885 (1987).
    • (1987) J. Am. Chem. Soc , vol.109 , pp. 6883-6885
    • Pettit, G.R.1
  • 180
    • 0029782186 scopus 로고    scopus 로고
    • Progress in the discovery of biosynthetic anticancer drugs
    • Pettit, G. R. Progress in the discovery of biosynthetic anticancer drugs. J. Nat. Prod. 59, 812-821 (1996).
    • (1996) J. Nat. Prod , vol.59 , pp. 812-821
    • Pettit, G.R.1
  • 181
    • 0024394570 scopus 로고
    • The absolute-configuration and synthesis of natural (-)-Dolastatin-10
    • Pettit, G. R. et al. The absolute-configuration and synthesis of natural (-)-Dolastatin-10. J. Am. Chem. Soc. 111, 5463-5465 (1989).
    • (1989) J. Am. Chem. Soc , vol.111 , pp. 5463-5465
    • Pettit, G.R.1
  • 182
    • 0025352537 scopus 로고
    • Dolastatin-10, a powerful cytostatic peptide derived from a marine animal - inhibition of tubulin polymerization mediated through the vinca alkaloid binding domain
    • Bai, R., Pettit, G. R. & Hamel, E. 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).
    • (1990) Biochem. Pharmacol , vol.39 , pp. 1941-1949
    • Bai, R.1    Pettit, G.R.2    Hamel, E.3
  • 183
    • 0025183762 scopus 로고
    • Binding of dolastatin-10 to tubulin at a distinct site for peptide antimitotic agents near the exchangeable nucleotide and vinca alkaloid sites
    • Bai, R., Pettit, G. R. & Hamel, E. 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).
    • (1990) J. Biol. Chem , vol.265 , pp. 17141-17149
    • Bai, R.1    Pettit, G.R.2    Hamel, E.3
  • 184
    • 0026748367 scopus 로고
    • Dolastatin-15, a potent antimitotic depsipeptide derived from Dolabella auricularia interaction with tubulin and effects on cellular microtubules
    • Bai, R., Friedman, S. J., Pettit, G. R. & Hamel, E. Dolastatin-15, a potent antimitotic depsipeptide derived from Dolabella auricularia interaction with tubulin and effects on cellular microtubules. Biochem. Pharmacol. 43, 2637-2645 (1992).
    • (1992) Biochem. Pharmacol , vol.43 , pp. 2637-2645
    • Bai, R.1    Friedman, S.J.2    Pettit, G.R.3    Hamel, E.4
  • 185
    • 0033960022 scopus 로고    scopus 로고
    • Sustained intracellular retention of dolastatin 10 causes its potent antimitotic activity
    • Verdier-Pinard, P., Kepler, J. A., Pettit, G. R. & Hamel, E. Sustained intracellular retention of dolastatin 10 causes its potent antimitotic activity. Mol. Pharmacol. 57, 180-187 (2000).
    • (2000) Mol. Pharmacol , vol.57 , pp. 180-187
    • Verdier-Pinard, P.1    Kepler, J.A.2    Pettit, G.R.3    Hamel, E.4
  • 186
    • 0033002547 scopus 로고    scopus 로고
    • Phase I trial of dolastatin-10 (NSC 376128) in patients with advanced solid tumors
    • Pitot, H. C. et al. Phase I trial of dolastatin-10 (NSC 376128) in patients with advanced solid tumors. Clin. Cancer Res. 5, 525-531 (1999).
    • (1999) Clin. Cancer Res , vol.5 , pp. 525-531
    • Pitot, H.C.1
  • 187
    • 0034005620 scopus 로고    scopus 로고
    • Novel marine-derived anticancer agents: A phase I clinical, pharmacological, and pharmacodynamic study of dolastatin 10 (NSC 376128) in patients with advanced solid tumors
    • Madden, T. et al. Novel marine-derived anticancer agents: A phase I clinical, pharmacological, and pharmacodynamic study of dolastatin 10 (NSC 376128) in patients with advanced solid tumors. Clin. Cancer Res. 6, 1293-1301 (2000).
    • (2000) Clin. Cancer Res , vol.6 , pp. 1293-1301
    • Madden, T.1
  • 188
    • 0033739893 scopus 로고    scopus 로고
    • Phase II study of dolastatin-10 in patients with hormone-refractory metastatic prostate adenocarcinoma
    • Vaishampayan, U. et al. Phase II study of dolastatin-10 in patients with hormone-refractory metastatic prostate adenocarcinoma. Clin. Cancer Res. 6, 4205-4208 (2000).
    • (2000) Clin. Cancer Res , vol.6 , pp. 4205-4208
    • Vaishampayan, U.1
  • 189
    • 0034093431 scopus 로고    scopus 로고
    • Phase II study of dolastatin-10 in patients with advanced non-small-cell lung cancer
    • Krug, L. M. et al. Phase II study of dolastatin-10 in patients with advanced non-small-cell lung cancer. Ann. Oncol. 11, 227-228 (2000).
    • (2000) Ann. Oncol , vol.11 , pp. 227-228
    • Krug, L.M.1
  • 190
    • 0034851434 scopus 로고    scopus 로고
    • Dolastatin-10 in metastatic melanoma: A phase II and pharmokinetic trial of the California Cancer Consortium
    • Margolin, K. et al. Dolastatin-10 in metastatic melanoma: A phase II and pharmokinetic trial of the California Cancer Consortium. Invest. New Drugs 19, 335-340 (2001).
    • (2001) Invest. New Drugs , vol.19 , pp. 335-340
    • Margolin, K.1
  • 191
    • 0036795186 scopus 로고    scopus 로고
    • Phase II study of dolastatin-10 as first-line treatment for advanced colorectal cancer
    • Saad, E. D. et al. Phase II study of dolastatin-10 as first-line treatment for advanced colorectal cancer. Am. J. Clin. Oncol. 25, 451-453 (2002).
    • (2002) Am. J. Clin. Oncol , vol.25 , pp. 451-453
    • Saad, E.D.1
  • 192
    • 11244283696 scopus 로고    scopus 로고
    • Phase II trial of dolastatin-10, a novel anti-tubulin agent, in metastatic soft tissue sarcomas
    • von Mehren, M. et al. Phase II trial of dolastatin-10, a novel anti-tubulin agent, in metastatic soft tissue sarcomas. Sarcoma 8, 107-111 (2004).
    • (2004) Sarcoma , vol.8 , pp. 107-111
    • von Mehren, M.1
  • 193
    • 18844461704 scopus 로고    scopus 로고
    • Phase II trial of dolastatin-10 in patients with advanced breast cancer
    • Perez, E. A. et al. Phase II trial of dolastatin-10 in patients with advanced breast cancer. Invest. New Drugs 23, 257-261 (2005).
    • (2005) Invest. New Drugs , vol.23 , pp. 257-261
    • Perez, E.A.1
  • 194
    • 23844535643 scopus 로고    scopus 로고
    • Phase II trials of dolastatin-10 in advanced pancreaticobiliary cancers
    • Kindler, H. L. et al. Phase II trials of dolastatin-10 in advanced pancreaticobiliary cancers. Invest. New Drugs 23, 489-493 (2005).
    • (2005) Invest. New Drugs , vol.23 , pp. 489-493
    • Kindler, H.L.1
  • 195
    • 4344650390 scopus 로고    scopus 로고
    • Marine natural products and related compounds in clinical and advanced preclinical trials
    • Newman, D. J. & Cragg, G. M. Marine natural products and related compounds in clinical and advanced preclinical trials. J. Nat. Prod. 67, 1216-1238 (2004).
    • (2004) J. Nat. Prod , vol.67 , pp. 1216-1238
    • Newman, D.J.1    Cragg, G.M.2
  • 196
    • 0030612225 scopus 로고    scopus 로고
    • Antitumor activity of TZT-1027, a novel dolastatin 10 derivative
    • Kobayashi, M. et al. Antitumor activity of TZT-1027, a novel dolastatin 10 derivative. Jpn. J. Cancer Res. 88, 316-327 (1997).
    • (1997) Jpn. J. Cancer Res , vol.88 , pp. 316-327
    • Kobayashi, M.1
  • 197
    • 18144432660 scopus 로고    scopus 로고
    • Phase I and pharmacokinetic study of TZT-1027, a novel synthetic dolastatin 10 derivative, administered as a 1-hour intravenous infusion every 3 weeks in patients with advanced refractory cancer
    • Schoffski, P. et al. Phase I and pharmacokinetic study of TZT-1027, a novel synthetic dolastatin 10 derivative, administered as a 1-hour intravenous infusion every 3 weeks in patients with advanced refractory cancer. Ann. Oncol. 15, 671-679 (2004).
    • (2004) Ann. Oncol , vol.15 , pp. 671-679
    • Schoffski, P.1
  • 198
    • 18844444472 scopus 로고    scopus 로고
    • Phase I and pharmacokinetic study of the dolastatin 10 analogue TZT-1027, given on days 1 and 8 of a 3-week cycle in patients with advanced solid tumors
    • de Jonge, M. J. et al. Phase I and pharmacokinetic study of the dolastatin 10 analogue TZT-1027, given on days 1 and 8 of a 3-week cycle in patients with advanced solid tumors. Clin. Cancer Res. 11 3806-3813 (2005).
    • (2005) Clin. Cancer Res , vol.11 , pp. 3806-3813
    • de Jonge, M.J.1
  • 199
    • 4444305445 scopus 로고    scopus 로고
    • TZT-1027 elucidates antitumor activity through direct cytotoxicity and selective blockade of blood supply
    • Hashiguchi, N. et al. TZT-1027 elucidates antitumor activity through direct cytotoxicity and selective blockade of blood supply. Anticancer Res. 24, 2201-2208 (2004).
    • (2004) Anticancer Res , vol.24 , pp. 2201-2208
    • Hashiguchi, N.1
  • 200
    • 0033853789 scopus 로고    scopus 로고
    • Characterization of the interaction of TZT-1027, a potent antitumor agent, with tubulin
    • Natsume, T. et al. Characterization of the interaction of TZT-1027, a potent antitumor agent, with tubulin. Jpn. J. Cancer Res. 91, 737-747 (2000).
    • (2000) Jpn. J. Cancer Res , vol.91 , pp. 737-747
    • Natsume, T.1
  • 201
    • 0022709534 scopus 로고
    • Halichondrins - antitumorpolyether macrolides from a marine sponge
    • Hirata, Y. & Uemura, D., Halichondrins - antitumorpolyether macrolides from a marine sponge. Pure App. Chem. 58, 701-710 (1986).
    • (1986) Pure App. Chem , vol.58 , pp. 701-710
    • Hirata, Y.1    Uemura, D.2
  • 202
    • 0022378694 scopus 로고
    • Norhalichondrin A: An antitumor polyether macrolide from a marine sponge
    • Uemura, D. et al. Norhalichondrin A: An antitumor polyether macrolide from a marine sponge. J. Am. Chem. Soc. 107, 4796-4798 (1985).
    • (1985) J. Am. Chem. Soc , vol.107 , pp. 4796-4798
    • Uemura, D.1
  • 203
    • 0027195860 scopus 로고
    • Isolation and structure of halistatin 1 from the eastern Indian Ocean marine sponge Phakellia carteri
    • Pettit, G. R. et al. Isolation and structure of halistatin 1 from the eastern Indian Ocean marine sponge Phakellia carteri. J. Org. Chem. 58, 2538-2543 (1993).
    • (1993) J. Org. Chem , vol.58 , pp. 2538-2543
    • Pettit, G.R.1
  • 204
    • 0026356420 scopus 로고
    • Antineoplastic agents. 219. Isolation and structure of the cell growth inhibitory constituents from the western Pacific marine sponge Axinella sp
    • Pettit, G. R. et al. Antineoplastic agents. 219. Isolation and structure of the cell growth inhibitory constituents from the western Pacific marine sponge Axinella sp. J. Med. Chem. 34, 3339-3340 (1991).
    • (1991) J. Med. Chem , vol.34 , pp. 3339-3340
    • Pettit, G.R.1
  • 205
    • 0026069885 scopus 로고
    • Halichondrin B and homohalichondrin B, marine natural products binding in the vinca domain of tubulin. Discovery of tubulin-based mechanism of action by analysis of differential cytotoxicity data
    • Bai, R. L. et al. Halichondrin B and homohalichondrin B, marine natural products binding in the vinca domain of tubulin. Discovery of tubulin-based mechanism of action by analysis of differential cytotoxicity data. J. Biol. Chem., 266, 15882-15889 (1991).
    • (1991) J. Biol. Chem , vol.266 , pp. 15882-15889
    • Bai, R.L.1
  • 206
    • 33751102419 scopus 로고    scopus 로고
    • Comparison of the activities of the truncated halichondrin B analog NSC 707389 (E7389) with those of the parent compound and a proposed binding site on tubulin
    • Dabydeen, D. A. et al. Comparison of the activities of the truncated halichondrin B analog NSC 707389 (E7389) with those of the parent compound and a proposed binding site on tubulin. Mol. Pharmacol. 70, 1866-1875 (2006).
    • (2006) Mol. Pharmacol , vol.70 , pp. 1866-1875
    • Dabydeen, D.A.1
  • 207
    • 0026774529 scopus 로고
    • Total synthesis of halichondrin B and norhalichondrin B
    • Aicher, T. D et al. Total synthesis of halichondrin B and norhalichondrin B. J. Am. Chem. Soc. 114, 3162-3164 (1992).
    • (1992) J. Am. Chem. Soc , vol.114 , pp. 3162-3164
    • Aicher, T.D.1
  • 209
    • 58149204512 scopus 로고    scopus 로고
    • Gravalos, D. G., Lake, R., Blunt, J. W., Munro, M. H. G. & Litaudon, M. S. P. Halichondrins: Cytotoxic polyether macrolides. EP0572109 (B1) (1993).
    • Gravalos, D. G., Lake, R., Blunt, J. W., Munro, M. H. G. & Litaudon, M. S. P. Halichondrins: Cytotoxic polyether macrolides. EP0572109 (B1) (1993).
  • 210
    • 0032993329 scopus 로고    scopus 로고
    • The discovery and development of marine compounds with pharmaceutical potential
    • Munro, M. H. G. et al. The discovery and development of marine compounds with pharmaceutical potential. J. Biotechnol. 70, 15-25 (1999).
    • (1999) J. Biotechnol , vol.70 , pp. 15-25
    • Munro, M.H.G.1
  • 211
    • 0001635334 scopus 로고    scopus 로고
    • New synthetic route to the C14-C38 segment of halichondrins
    • Stamos, D. P., Sean, S. C. & Kishi, Y. New synthetic route to the C14-C38 segment of halichondrins. J. Org. Chem. 62, 7552-7553 (1997).
    • (1997) J. Org. Chem , vol.62 , pp. 7552-7553
    • Stamos, D.P.1    Sean, S.C.2    Kishi, Y.3
  • 213
    • 58149181807 scopus 로고    scopus 로고
    • Littlefield, B. A. et al. Macrocyclic analogs and methods of their use and preparation. WO9965894 (A1) (1999).
    • Littlefield, B. A. et al. Macrocyclic analogs and methods of their use and preparation. WO9965894 (A1) (1999).
  • 214
    • 84975897156 scopus 로고    scopus 로고
    • eds Cragg, G. M, Kingston, D. G. I. & Newman, D. J, Taylor and Francis, Boca Raton
    • Yu, M. J., Kishi, Y. & Littlefield, B. A. in Anticancer Agents from Natural Products (eds Cragg, G. M., Kingston, D. G. I. & Newman, D. J.) 241-265 (Taylor and Francis, Boca Raton, 2005).
    • (2005) Anticancer Agents from Natural Products , pp. 241-265
    • Yu, M.J.1    Kishi, Y.2    Littlefield, B.A.3
  • 215
    • 0035110756 scopus 로고    scopus 로고
    • In vitro and in vivo anticancer activities of synthetic macrocyclic ketone analogues of halichondrin B
    • Towle, M. J. et al. In vitro and in vivo anticancer activities of synthetic macrocyclic ketone analogues of halichondrin B. Cancer Res. 61, 1013-1021 (2001).
    • (2001) Cancer Res , vol.61 , pp. 1013-1021
    • Towle, M.J.1
  • 216
    • 33750359578 scopus 로고    scopus 로고
    • E7389, a novel anti-tubulin, in patients with refractory breast cancer
    • Forero, J. B. et al. E7389, a novel anti-tubulin, in patients with refractory breast cancer. J. Clin. Oncol. 24 (Suppl. 18), 653 (2006).
    • (2006) J. Clin. Oncol , vol.24 , Issue.SUPPL. 18 , pp. 653
    • Forero, J.B.1
  • 217
    • 47549105681 scopus 로고    scopus 로고
    • Phase II study of eribulin mesylate (E7389) halichondrin B analog in patients with refractory breast cancer
    • Blum, J. L. et al. Phase II study of eribulin mesylate (E7389) halichondrin B analog in patients with refractory breast cancer. J. Clin. Oncol. 25 (Suppl. 18), 1034 (2007).
    • (2007) J. Clin. Oncol , vol.25 , Issue.SUPPL. 18 , pp. 1034
    • Blum, J.L.1
  • 218
    • 45349107525 scopus 로고    scopus 로고
    • Phase II study of eribulin mesylate (E7389), a mechanistically novel inhibitor of microtubule dynamics, in patients with advanced non-small cell lung cancer (NSCLC)
    • Spira, A. I., et al. Phase II study of eribulin mesylate (E7389), a mechanistically novel inhibitor of microtubule dynamics, in patients with advanced non-small cell lung cancer (NSCLC). J. Clin. Oncol. 25 (Suppl. 18), 7546 (2007).
    • (2007) J. Clin. Oncol , vol.25 , Issue.SUPPL. 18 , pp. 7546
    • Spira, A.I.1
  • 219
    • 58149203665 scopus 로고    scopus 로고
    • PR Newswire. Eribulin Mesylate Demonstrated Anti-Tumor Activity in Heavily Pretreated Patients With Advanced Breast Cancer. PR Newswire web site [online], (2008).
    • PR Newswire. Eribulin Mesylate Demonstrated Anti-Tumor Activity in Heavily Pretreated Patients With Advanced Breast Cancer. PR Newswire web site [online], (2008).
  • 220
    • 0027296890 scopus 로고    scopus 로고
    • Hamann, M. T. & Scheuer, P. J. 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).
    • Hamann, M. T. & Scheuer, P. J. 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).
  • 221
    • 0033580898 scopus 로고    scopus 로고
    • The absolute stereochemistry of kahalalide F
    • Goetz, G., Yoshida, W. Y. & Scheuer, P. J. The absolute stereochemistry of kahalalide F. Tetrahedron 55, 7739-7746 (1999).
    • (1999) Tetrahedron , vol.55 , pp. 7739-7746
    • Goetz, G.1    Yoshida, W.Y.2    Scheuer, P.J.3
  • 222
    • 0029842550 scopus 로고    scopus 로고
    • Kahalalides: Bioactive peptide from a marine mollusk Elysia rufescens and its algal diet Bryopsis sp
    • Hamann, M. T., Otto, C. S., Scheuer, P. J. & Dunbar, D. C. Kahalalides: bioactive peptide from a marine mollusk Elysia rufescens and its algal diet Bryopsis sp. J. Org. Chem. 61, 6594-6600 (1996).
    • (1996) J. Org. Chem , vol.61 , pp. 6594-6600
    • Hamann, M.T.1    Otto, C.S.2    Scheuer, P.J.3    Dunbar, D.C.4
  • 223
    • 0027944430 scopus 로고
    • Structure of cypemycin, a new peptide antibiotic
    • Minami, Y. et al. Structure of cypemycin, a new peptide antibiotic. Tetrahedron Lett. 35, 8001-8004 (1994).
    • (1994) Tetrahedron Lett , vol.35 , pp. 8001-8004
    • Minami, Y.1
  • 224
    • 20444448150 scopus 로고    scopus 로고
    • Hassallidin A, a glycosylated lipopeptide with antifungal activity from the cyanobacterium Hassallia sp
    • Neuhof, T. et al. Hassallidin A, a glycosylated lipopeptide with antifungal activity from the cyanobacterium Hassallia sp. J. Nat. Prod. 68, 695-700 (2005).
    • (2005) J. Nat. Prod , vol.68 , pp. 695-700
    • Neuhof, T.1
  • 225
    • 0035161213 scopus 로고    scopus 로고
    • Chemical defenses of the sacoglossan mollusk Elysia rufescens and its host alga Bryopsis sp
    • Becerro, M. A., Goetz, G., Paul, V. J. & Scheuer, P. J. Chemical defenses of the sacoglossan mollusk Elysia rufescens and its host alga Bryopsis sp. J. Chem. Ecol. 27, 2287-2299 (2001).
    • (2001) J. Chem. Ecol , vol.27 , pp. 2287-2299
    • Becerro, M.A.1    Goetz, G.2    Paul, V.J.3    Scheuer, P.J.4
  • 228
    • 0030053875 scopus 로고    scopus 로고
    • The antitumoral compound Kahalalide F acts on cell lysosomes
    • Garcia-Rocha, M., Bonay, P. & Avila, J. The antitumoral compound Kahalalide F acts on cell lysosomes. Cancer Lett. 99, 43-50 (1996).
    • (1996) Cancer Lett , vol.99 , pp. 43-50
    • Garcia-Rocha, M.1    Bonay, P.2    Avila, J.3
  • 229
    • 0642318239 scopus 로고    scopus 로고
    • Kahalalide F. a new marine-derived compound, induces oncosis in human prostate and breast cancer cells
    • Suarez, Y. et al. Kahalalide F. a new marine-derived compound, induces oncosis in human prostate and breast cancer cells, Mol. Cancer Ther. 2, 863-872 (2003).
    • (2003) Mol. Cancer Ther , vol.2 , pp. 863-872
    • Suarez, Y.1
  • 230
    • 23044479629 scopus 로고    scopus 로고
    • Kahalalide F induces necrosis-like cell death that involves depletion of ErbB3 and inhibition of Akt signaling
    • Janmaat, M. L., Rodriguez, J. A., Jimeno, J., Kruyt, F. A. E. & Giaccone, G. Kahalalide F induces necrosis-like cell death that involves depletion of ErbB3 and inhibition of Akt signaling. Mol. Pharmocol. 68, 502-510 (2005).
    • (2005) Mol. Pharmocol , vol.68 , pp. 502-510
    • Janmaat, M.L.1    Rodriguez, J.A.2    Jimeno, J.3    Kruyt, F.A.E.4    Giaccone, G.5
  • 231
    • 22144492575 scopus 로고    scopus 로고
    • The mechanism of action of Kahalalide F: Variable cell permeability in human hepatoma cell lines
    • Sewell, J. M. et al. The mechanism of action of Kahalalide F: variable cell permeability in human hepatoma cell lines. Eur J. Cancer 41, 1637-1644 (2005).
    • (2005) Eur J. Cancer , vol.41 , pp. 1637-1644
    • Sewell, J.M.1
  • 232
    • 0034887240 scopus 로고    scopus 로고
    • Chemical and enzymatic stability of a cyclic depsipeptide, the novel, marine-derived, anti-cancer agent kahalalide F
    • Sparidans, R. W. et al. Chemical and enzymatic stability of a cyclic depsipeptide, the novel, marine-derived, anti-cancer agent kahalalide F. Anticancer Drugs 12, 575-582 (2001).
    • (2001) Anticancer Drugs , vol.12 , pp. 575-582
    • Sparidans, R.W.1
  • 233
    • 0036392375 scopus 로고    scopus 로고
    • Preclinical toxicity studies of kahalalide F, a new anticancer agent: Single and multiple dosing regimens in the rat
    • Brown, A. P., Morrissey R. L., Faircloth, G. T. & Levine, B. S. Preclinical toxicity studies of kahalalide F, a new anticancer agent: single and multiple dosing regimens in the rat. Cancer Chemother Pharmacol. 50, 333-340 (2002).
    • (2002) Cancer Chemother Pharmacol , vol.50 , pp. 333-340
    • Brown, A.P.1    Morrissey, R.L.2    Faircloth, G.T.3    Levine, B.S.4
  • 234
    • 20144387163 scopus 로고    scopus 로고
    • Phase I clinical and pharmacokinetic study of Kahalalide F in patients with advanced androgen refractory prostate cancer
    • Rademaker-Lakhai, J. M. et al. Phase I clinical and pharmacokinetic study of Kahalalide F in patients with advanced androgen refractory prostate cancer. Clin. Cancer Res. 11, 1854-1862 (2005).
    • (2005) Clin. Cancer Res , vol.11 , pp. 1854-1862
    • Rademaker-Lakhai, J.M.1
  • 235
    • 0042458241 scopus 로고    scopus 로고
    • A phase I clinical and pharmacokinetic (PK) study with Kahalalide F (KF) in patients (pts) with advanced solid tumors (AST) with a continuous weekly W 1-hour iv infusion schedule
    • S
    • Ciruelos, C. et al. A phase I clinical and pharmacokinetic (PK) study with Kahalalide F (KF) in patients (pts) with advanced solid tumors (AST) with a continuous weekly W 1-hour iv infusion schedule. Eur.J. Cancer 38 (Suppl.), S33 (2002).
    • (2002) Eur.J. Cancer , vol.38 , Issue.SUPPL. , pp. 33
    • Ciruelos, C.1
  • 236
    • 0020408957 scopus 로고
    • Isolation and structure of bryostatin-1
    • Pettit, G. R. et al. Isolation and structure of bryostatin-1 J. Am. Chem. Soc. 104, 6846-6848 (1982).
    • (1982) J. Am. Chem. Soc , vol.104 , pp. 6846-6848
    • Pettit, G.R.1
  • 237
    • 0026093899 scopus 로고
    • The large-scale isolation of bryostatin-1 from Bugula neritina following current good manufacturing practices
    • Schaufelberger, D. E. et al. The large-scale isolation of bryostatin-1 from Bugula neritina following current good manufacturing practices. J Nat. Prod. 54, 1265-1270 (1991).
    • (1991) J Nat. Prod , vol.54 , pp. 1265-1270
    • Schaufelberger, D.E.1
  • 238
    • 33751156706 scopus 로고
    • Supply issues complicate trek of chemicals from sea to market
    • Rouhi, M. A. Supply issues complicate trek of chemicals from sea to market. Chem. Eng. News 73, 42-44 (1995).
    • (1995) Chem. Eng. News , vol.73 , pp. 42-44
    • Rouhi, M.A.1
  • 239
    • 0033603858 scopus 로고    scopus 로고
    • Total synthesis of bryostatin 2
    • Evans, D. A. et al. Total synthesis of bryostatin 2. J. Am. Chem. Soc. 121, 7540-7552 (1999).
    • (1999) J. Am. Chem. Soc , vol.121 , pp. 7540-7552
    • Evans, D.A.1
  • 240
    • 0034600771 scopus 로고    scopus 로고
    • Total synthesis of bryostatin 3
    • Ohmori, K. et al. Total synthesis of bryostatin 3. Angew. Chem. Int. Ed. Engl. 39, 2290-2294 (2000).
    • (2000) Angew. Chem. Int. Ed. Engl , vol.39 , pp. 2290-2294
    • Ohmori, K.1
  • 241
    • 2642554823 scopus 로고    scopus 로고
    • Evolution of synthetic strategies for highly functionalized natural products: A successful route to bryostatin 3
    • Ohmori, K. Evolution of synthetic strategies for highly functionalized natural products: A successful route to bryostatin 3. Bull. Chem. Soc. Jap. 77, 875-885 (2004).
    • (2004) Bull. Chem. Soc. Jap , vol.77 , pp. 875-885
    • Ohmori, K.1
  • 242
    • 0025053566 scopus 로고
    • Synthesis of bryostatin-7
    • Kageyama, M. et al. Synthesis of bryostatin-7. J. Am. Chem. Soc. 112, 7407-7408 (1990).
    • (1990) J. Am. Chem. Soc , vol.112 , pp. 7407-7408
    • Kageyama, M.1
  • 243
    • 0024406910 scopus 로고
    • Synthesis of bryostatins. 1. construction of the C(1)-C(16) fragment
    • Blanchette, M. A. et al. Synthesis of bryostatins. 1. construction of the C(1)-C(16) fragment. J. Org. Chem. 54, 2817-2825 (1989).
    • (1989) J. Org. Chem , vol.54 , pp. 2817-2825
    • Blanchette, M.A.1
  • 244
    • 8744304742 scopus 로고    scopus 로고
    • Synthesis of the bryostatin 1 northern hemisphere (C1-C16) via desymmetrization by ketalization/ring-closing metathesis
    • Voight, E. A., Seradi, H., Roethle, P. A. & Burke, S. D. Synthesis of the bryostatin 1 northern hemisphere (C1-C16) via desymmetrization by ketalization/ring-closing metathesis. Org. Lett. 6, 4045-4048 (2004).
    • (2004) Org. Lett , vol.6 , pp. 4045-4048
    • Voight, E.A.1    Seradi, H.2    Roethle, P.A.3    Burke, S.D.4
  • 245
    • 0000746887 scopus 로고    scopus 로고
    • Towards the asymmetric synthesis of bryostatin 1
    • DeBrabander, J. & Vandewalle, M. Towards the asymmetric synthesis of bryostatin 1. Pure Appl. Chem. 68, 715-718 (1996).
    • (1996) Pure Appl. Chem , vol.68 , pp. 715-718
    • DeBrabander, J.1    Vandewalle, M.2
  • 246
    • 0027322773 scopus 로고
    • Inhibition of phorbol ester-induced T-cell proliferation by bryostatin is associated with rapid degradation of protein-kinase-C
    • Isakov, N., Galron, D., Mustelin, T, Pettit, G. R. & Altman, A. Inhibition of phorbol ester-induced T-cell proliferation by bryostatin is associated with rapid degradation of protein-kinase-C. J. Immunol. 150, 1195-1204 (1993).
    • (1993) J. Immunol , vol.150 , pp. 1195-1204
    • Isakov, N.1    Galron, D.2    Mustelin, T.3    Pettit, G.R.4    Altman, A.5
  • 247
    • 0032513707 scopus 로고    scopus 로고
    • Synthesis of the first members of a new class of biologically active bryostatin analogues
    • Wender, P. A. et al. Synthesis of the first members of a new class of biologically active bryostatin analogues. J. Am. Chem. Soc. 120, 4534-4535 (1998).
    • (1998) J. Am. Chem. Soc , vol.120 , pp. 4534-4535
    • Wender, P.A.1
  • 248
    • 0000852678 scopus 로고
    • Modeling of the bryostatins to the phorbol ester pharmacophore on protein kinase
    • Wender, P. A. et al. Modeling of the bryostatins to the phorbol ester pharmacophore on protein kinase C. Proc. Natl Acod. Sci. USA 85, 7197-7201 (1988).
    • (1988) C. Proc. Natl Acod. Sci. USA , vol.85 , pp. 7197-7201
    • Wender, P.A.1
  • 249
    • 0023253596 scopus 로고
    • Bryostatin 1. an activator of protein kinase C, inhibits tumor promotion by phorbol esters in SENCAR mouse skin
    • Hennings, H. et al. Bryostatin 1. an activator of protein kinase C, inhibits tumor promotion by phorbol esters in SENCAR mouse skin. Carcinogenesis 8, 1343-1346 (1987).
    • (1987) Carcinogenesis , vol.8 , pp. 1343-1346
    • Hennings, H.1
  • 250
    • 0027315073 scopus 로고
    • In vivo administration of the anticancer agent bryostatin 1 activates platelets and neutrophils and modulates protein kinase C activity
    • Berkow, R. L. et al. In vivo administration of the anticancer agent bryostatin 1 activates platelets and neutrophils and modulates protein kinase C activity. Cancer Res. 53, 2810-2815 (1993).
    • (1993) Cancer Res , vol.53 , pp. 2810-2815
    • Berkow, R.L.1
  • 251
    • 0027436897 scopus 로고
    • Modulation of gene-expression in the acute promyelocytic leukemia-cell line Nb4
    • Hu, Z. B., Ma, W. L., Uphoff, C. C., Lanotte, M. & Drexler, H. G. Modulation of gene-expression in the acute promyelocytic leukemia-cell line Nb4. Leukemia 7, 1817-1823 (1993).
    • (1993) Leukemia , vol.7 , pp. 1817-1823
    • Hu, Z.B.1    Ma, W.L.2    Uphoff, C.C.3    Lanotte, M.4    Drexler, H.G.5
  • 252
    • 0026531518 scopus 로고
    • Effect of bryostatin-1 on the in vitro radioprotective capacity of recombinant granulocyte macrophage colony-stimulating factor (Rgm-Csf) toward committed human myeloid progenitor cells (Cfu-Gm)
    • Grant, S., Pettit, G. R. & McCrady, C. Effect of bryostatin-1 on the in vitro radioprotective capacity of recombinant granulocyte macrophage colony-stimulating factor (Rgm-Csf) toward committed human myeloid progenitor cells (Cfu-Gm). Exp. Hematol. 20, 34-42 (1992).
    • (1992) Exp. Hematol , vol.20 , pp. 34-42
    • Grant, S.1    Pettit, G.R.2    McCrady, C.3
  • 253
    • 0033868507 scopus 로고    scopus 로고
    • Chemistry and clinical biology of the bryostatins
    • Mutter, R. & Wills, M. Chemistry and clinical biology of the bryostatins. Bioorg. Med. Chem. 8, 1841-1860 (2000).
    • (2000) Bioorg. Med. Chem , vol.8 , pp. 1841-1860
    • Mutter, R.1    Wills, M.2
  • 254
    • 0031760304 scopus 로고    scopus 로고
    • A phase II study of bryostatin 1 in metastatic malignant melanoma
    • Propper, D. J. et al. A phase II study of bryostatin 1 in metastatic malignant melanoma. Br. J. Cancer 78, 1337-1341 (1998).
    • (1998) Br. J. Cancer , vol.78 , pp. 1337-1341
    • Propper, D.J.1
  • 255
    • 0344623689 scopus 로고    scopus 로고
    • Phase I tríal of bryostatin I in relapsed lymphoma and CLL
    • Varterasian, M. et al. Phase I tríal of bryostatin I in relapsed lymphoma and CLL. Blood 88, 2269 (1996).
    • (1996) Blood , vol.88 , pp. 2269
    • Varterasian, M.1
  • 256
    • 0034851019 scopus 로고    scopus 로고
    • Phase II study of bryostatin 1 in patients with relapsed multiple myeloma
    • Varterasian, M. L. et al. Phase II study of bryostatin 1 in patients with relapsed multiple myeloma. Invest. New Drugs 19 245-247 (2001).
    • (2001) Invest. New Drugs , vol.19 , pp. 245-247
    • Varterasian, M.L.1
  • 257
    • 12944249449 scopus 로고    scopus 로고
    • Phase II trial of bryostatin 1 in patients with relapsed low-grade non-Hodgkin's lymphoma and chronic lymphocytic leukemia
    • Varterasian, M. L. et al. Phase II trial of bryostatin 1 in patients with relapsed low-grade non-Hodgkin's lymphoma and chronic lymphocytic leukemia Clin. Cancer Res. 6, 825-828 (2000).
    • (2000) Clin. Cancer Res , vol.6 , pp. 825-828
    • Varterasian, M.L.1
  • 258
    • 0031954529 scopus 로고    scopus 로고
    • Phase lb trial of bryostatin 1 in patients with refractory malignancies
    • Grant, S. et al. Phase lb trial of bryostatin 1 in patients with refractory malignancies. Clin. Cancer Res. 4, 611-618 (1998).
    • (1998) Clin. Cancer Res , vol.4 , pp. 611-618
    • Grant, S.1
  • 259
    • 0036667939 scopus 로고    scopus 로고
    • The clinical development of the bryostatins
    • Clamp, A. & Jayson, G. C. The clinical development of the bryostatins, Anticancer Drugs 13, 673-683 (2002).
    • (2002) Anticancer Drugs , vol.13 , pp. 673-683
    • Clamp, A.1    Jayson, G.C.2
  • 260
    • 0028787625 scopus 로고
    • Bryostatin 1, a novel antineoplastic agent and protein-kinase-C activator, induces human myalgia and muscle metabolic defects - a P-31 magnetic-resonance spectroscopic study
    • Hickman, P. F. et al. Bryostatin 1, a novel antineoplastic agent and protein-kinase-C activator, induces human myalgia and muscle metabolic defects - a P-31 magnetic-resonance spectroscopic study. Br. J. Cancer 72, 998-1003 (1995).
    • (1995) Br. J. Cancer , vol.72 , pp. 998-1003
    • Hickman, P.F.1
  • 261
    • 0036210351 scopus 로고    scopus 로고
    • A phase II trial of bryostatin-1 in patients with metastatic or recurrent squamous cell carcinoma of the head and neck
    • Pfister, D. G. et al. A phase II trial of bryostatin-1 in patients with metastatic or recurrent squamous cell carcinoma of the head and neck. Invest. New Drugs 20, 123-127 (2002).
    • (2002) Invest. New Drugs , vol.20 , pp. 123-127
    • Pfister, D.G.1
  • 262
    • 0035139097 scopus 로고    scopus 로고
    • A phase II trial of bryostatin 1 in the treatment of metastatic colorectal cancer
    • Zonder, J. A. et al. A phase II trial of bryostatin 1 in the treatment of metastatic colorectal cancer. Clin, Cancer Res. 7 38-42 (2001).
    • (2001) Clin, Cancer Res , vol.7 , pp. 38-42
    • Zonder, J.A.1
  • 263
    • 0035035103 scopus 로고    scopus 로고
    • Phase II evaluation of bryostatin-1 in metastatic melanoma
    • Bedikian, A. Y. et al. Phase II evaluation of bryostatin-1 in metastatic melanoma. Melanoma Res. 11, 183-188 (2001).
    • (2001) Melanoma Res , vol.11 , pp. 183-188
    • Bedikian, A.Y.1
  • 264
    • 0034254971 scopus 로고    scopus 로고
    • A phase II trial of bryostatin-1 for patients with metastatic renal cell carcinoma
    • Pagliaro, L et al. A phase II trial of bryostatin-1 for patients with metastatic renal cell carcinoma. Cancer 89, 615-618 (2000).
    • (2000) Cancer , vol.89 , pp. 615-618
    • Pagliaro, L.1
  • 265
    • 0242298228 scopus 로고    scopus 로고
    • A phase II trial of bryostatin-I administered by weekly 24-hour infusion in recurrent epithelial ovarian carcinoma
    • Clamp, A. R. et al. A phase II trial of bryostatin-I administered by weekly 24-hour infusion in recurrent epithelial ovarian carcinoma. Br. J. Cancer 89, 1152-1154 (2003).
    • (2003) Br. J. Cancer , vol.89 , pp. 1152-1154
    • Clamp, A.R.1
  • 266
    • 0035992301 scopus 로고    scopus 로고
    • Phase I trial and correlative laboratory studies of bryostatin I (NSC 339555) and high-dose -β-D-arabinofuranosylcytosine in patients with refractory acute leukemia
    • Cragg, L. H. et al. Phase I trial and correlative laboratory studies of bryostatin I (NSC 339555) and high-dose -β-D-arabinofuranosylcytosine in patients with refractory acute leukemia. Clin. Cancer Res. 8, 2123-2133 (2002).
    • (2002) Clin. Cancer Res , vol.8 , pp. 2123-2133
    • Cragg, L.H.1
  • 267
    • 0347995049 scopus 로고    scopus 로고
    • Phase I and correlative study of combination bryostatin I and vincristine in relapsed B-cell malignancies
    • Dowlati, A. et al. Phase I and correlative study of combination bryostatin I and vincristine in relapsed B-cell malignancies. Clin. Cancer Res. 9, 5929-5935 (2003).
    • (2003) Clin. Cancer Res , vol.9 , pp. 5929-5935
    • Dowlati, A.1
  • 268
    • 33646501280 scopus 로고    scopus 로고
    • Multi-center Phase II study of sequential paclitaxel and bryostatin-1 (NSC 339555) in patients with untreated, advanced gastric or gastroesophageal junction adenocarcinoma
    • Ajani, J. A. Multi-center Phase II study of sequential paclitaxel and bryostatin-1 (NSC 339555) in patients with untreated, advanced gastric or gastroesophageal junction adenocarcinoma. Invest. New Drugs 24, 353-357 (2006).
    • (2006) Invest. New Drugs , vol.24 , pp. 353-357
    • Ajani, J.A.1
  • 269
    • 33644830944 scopus 로고    scopus 로고
    • Targeting the cell cycle: A new approach to cancer therapy
    • Schwartz, G. K. & Shah, M. A. Targeting the cell cycle: A new approach to cancer therapy. J. Clin. Oncol. 23, 9408-9421 (2005).
    • (2005) J. Clin. Oncol , vol.23 , pp. 9408-9421
    • Schwartz, G.K.1    Shah, M.A.2
  • 270
    • 0037208308 scopus 로고    scopus 로고
    • Property distributions: Differences between drugs, natural products, and molecules from combinatorial chemistry
    • Feher, M. & Schmidt, J. M. Property distributions: Differences between drugs, natural products, and molecules from combinatorial chemistry. J. Chem. Inf. Comput. Sci. 43, 218-227(2003).
    • (2003) J. Chem. Inf. Comput. Sci , vol.43 , pp. 218-227
    • Feher, M.1    Schmidt, J.M.2
  • 271
    • 34447302758 scopus 로고    scopus 로고
    • Udwary, D. W. et al. Genome sequencing reveals complex secondary metabolome in the marine actinomycete Salinispora tropico. Proc. Natl Acad. Sci. USA 104, 10376-10381 (2007).
    • Udwary, D. W. et al. Genome sequencing reveals complex secondary metabolome in the marine actinomycete Salinispora tropico. Proc. Natl Acad. Sci. USA 104, 10376-10381 (2007).
  • 272
    • 33745045183 scopus 로고    scopus 로고
    • New drugs from marine microbes: The tide is turning
    • Newman, D. J. & Hill, R. T. New drugs from marine microbes: The tide is turning. J. Ind. Microbiol. Biotechnol. 33, 539-544 (2006).
    • (2006) J. Ind. Microbiol. Biotechnol , vol.33 , pp. 539-544
    • Newman, D.J.1    Hill, R.T.2
  • 273
    • 85047689741 scopus 로고    scopus 로고
    • Bacterial symbionts; prospects for the sustainable production of invertebrate-derived pharmaceuticals
    • Piel, J. Bacterial symbionts; prospects for the sustainable production of invertebrate-derived pharmaceuticals. Curr. Med. Chem. 13 39-50 (2006).
    • (2006) Curr. Med. Chem , vol.13 , pp. 39-50
    • Piel, J.1
  • 274
    • 8844270242 scopus 로고    scopus 로고
    • bryA: An unusual modular polyketide synthase gene from the uncultivated bacterial symbiont of the marine bryozoan Bugula neritina
    • Hildebrand, M. et al. bryA: An unusual modular polyketide synthase gene from the uncultivated bacterial symbiont of the marine bryozoan Bugula neritina. Chem. Biol. 11, 1543-1552 (2004).
    • (2004) Chem. Biol , vol.11 , pp. 1543-1552
    • Hildebrand, M.1
  • 275
    • 33847029508 scopus 로고    scopus 로고
    • Identification of the putative bryostatin polyketide synthase gene cluster from "Candidatus Endobugula sertula", the uncultivated microbial symbiont from the marine bryozoan Bugula neritina
    • Sudek, S. et al. Identification of the putative bryostatin polyketide synthase gene cluster from "Candidatus Endobugula sertula", the uncultivated microbial symbiont from the marine bryozoan Bugula neritina. J. Nat. Prod. 70, 67-74 (2007).
    • (2007) J. Nat. Prod , vol.70 , pp. 67-74
    • Sudek, S.1
  • 277
    • 58149180418 scopus 로고    scopus 로고
    • Homer, The Odyssey (∼ 7th century B. C). Fagles, R. (translation). Penguin Books (2006).
    • Homer, The Odyssey (∼ 7th century B. C). Fagles, R. (translation). Penguin Books (2006).
  • 278
    • 0037455147 scopus 로고    scopus 로고
    • Salinosporamide A: A highly cytotoxic proteasome inhibitor from a novel microbial source, a marine bacterium of the new genus Salinospora
    • Feling, R. H. et al. Salinosporamide A: A highly cytotoxic proteasome inhibitor from a novel microbial source, a marine bacterium of the new genus Salinospora. Angew. Chem. Ind. Ed. 42, 355-357 (2003).
    • (2003) Angew. Chem. Ind. Ed , vol.42 , pp. 355-357
    • Feling, R.H.1
  • 279
    • 20444459450 scopus 로고    scopus 로고
    • Maldonado, L., Fenical, W., Goodfellow, M., Jensen, P. R. & Ward, A. C. Salinispora gen nov., sp. nov., Salinispora arenicola sp. nov., and S. tropica sp. nov., obligate marine actinomycetes belonging to the family Micromonosporaceae. Internat. J. System. Appl. Microbiol. 55, 1759-1766 (2005).
    • Maldonado, L., Fenical, W., Goodfellow, M., Jensen, P. R. & Ward, A. C. Salinispora gen nov., sp. nov., Salinispora arenicola sp. nov., and S. tropica sp. nov., obligate marine actinomycetes belonging to the family Micromonosporaceae. Internat. J. System. Appl. Microbiol. 55, 1759-1766 (2005).
  • 280
    • 27644562277 scopus 로고    scopus 로고
    • A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from Bortezomib
    • Chauhan, D. et al. A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from Bortezomib. Cancer Cell 8, 407-419 (2005).
    • (2005) Cancer Cell , vol.8 , pp. 407-419
    • Chauhan, D.1
  • 281
    • 38949125853 scopus 로고    scopus 로고
    • Combination of proteasome inhibitors bortezomib and NPI-0052 trigger in vivo synergistic cytotoxicity in multiple myeloma
    • Chauhan, D. et al. Combination of proteasome inhibitors bortezomib and NPI-0052 trigger in vivo synergistic cytotoxicity in multiple myeloma. Blood 111, 1654-1664 (2008).
    • (2008) Blood , vol.111 , pp. 1654-1664
    • Chauhan, D.1
  • 282
    • 62149103377 scopus 로고    scopus 로고
    • Discovery and development of the anticancer agent salinosporamide A (NPI-0052)
    • 5 Nov, doi:10.1016/j. bmc.2008.10.075
    • Fenicala, W. et al. Discovery and development of the anticancer agent salinosporamide A (NPI-0052). Bioorg. Med. Chem. 5 Nov 2008 (doi:10.1016/j. bmc.2008.10.075).
    • (2008) Bioorg. Med. Chem
    • Fenicala, W.1
  • 283
    • 0029100620 scopus 로고
    • Phorboxazoles A and B - potent cytostatic macrolides from marine sponge Phorbas sp
    • Searle, P. A. & Molinski, T. F. Phorboxazoles A and B - potent cytostatic macrolides from marine sponge Phorbas sp. J. Am. Chem. Soc. 117, 8126-8131 (1995).
    • (1995) J. Am. Chem. Soc , vol.117 , pp. 8126-8131
    • Searle, P.A.1    Molinski, T.F.2
  • 284
    • 0029854666 scopus 로고    scopus 로고
    • Absolute configuration of phorboxazoles A and B from the marine sponge Phorbas sp.1. macrolide and hemiketal rings
    • Searle, P. A., Molinski, T. F., Brzezinski, L. J. & Leahy, J. W. Absolute configuration of phorboxazoles A and B from the marine sponge Phorbas sp.1. macrolide and hemiketal rings. J. Am. Chem. Soc. 118, 9422-9423 (1996).
    • (1996) J. Am. Chem. Soc , vol.118 , pp. 9422-9423
    • Searle, P.A.1    Molinski, T.F.2    Brzezinski, L.J.3    Leahy, J.W.4
  • 285
    • 0030605181 scopus 로고    scopus 로고
    • Absolute configuration of phorboxazoles A and B from the marine sponge, Phorbas sp.2. C43 and complete stereochemistry
    • Molinski T. F. Absolute configuration of phorboxazoles A and B from the marine sponge, Phorbas sp.2. C43 and complete stereochemistry. Tetrahedron Lett. 37, 7879-7880 (1996).
    • (1996) Tetrahedron Lett , vol.37 , pp. 7879-7880
    • Molinski, T.F.1
  • 286
    • 34247112862 scopus 로고    scopus 로고
    • Skepper, C. K., MacMillan, J. B., Zhou, G.-X., Masuno, M. N. & Molinski, T. F. Chlorocyclopropane macrolides from the marine sponge Phorbas sp. Assignment of the absolute configurations of phorbasides A and B by quantitative CD. J. Am. Chem. Soc. 129, 4150-4151 (2007).
    • Skepper, C. K., MacMillan, J. B., Zhou, G.-X., Masuno, M. N. & Molinski, T. F. Chlorocyclopropane macrolides from the marine sponge Phorbas sp. Assignment of the absolute configurations of phorbasides A and B by quantitative CD. J. Am. Chem. Soc. 129, 4150-4151 (2007).
  • 287
    • 43449108881 scopus 로고    scopus 로고
    • Phorbasides A-E, cytotoxic chlorocyclopropane macrolide glycosides from the marine sponge Phorbas sp. CD determination of C-methyl sugar configurations
    • MacMillan, J. B., Xiong-Zhou, G., Skepper, C. K. & Molinski, T. F. Phorbasides A-E, cytotoxic chlorocyclopropane macrolide glycosides from the marine sponge Phorbas sp. CD determination of C-methyl sugar configurations. J. Org. Chem. 73, 3699-3706 (2008).
    • (2008) J. Org. Chem , vol.73 , pp. 3699-3706
    • MacMillan, J.B.1    Xiong-Zhou, G.2    Skepper, C.K.3    Molinski, T.F.4


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.