#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Active substances from marine organisms in clinical trials and practice


Authors: D. Mareček;  PharmDr. Jana Rudá-Kučerová, PhD.
Published in: Čes. slov. Farm., 2017; 66, 191-207
Category: Review Articles

Overview

Oceans cover a large part of our planet and they are a home for an enormous amount of species. A lot of them are still waiting to be discovered by man, much like the chemicals they synthesize. Marine pharmacology concerns itself with the study of these chemicals and their potential use in medicine. Origin in marine species is for the most part the only thing this large and diverse group of substances have in common, so the spectrum of possible applications is quite wide. Many of these substances have a unique mechanism of action, offering new therapeutic possibilities. Although just a few of them are used in a clinical practice today (e.g. eribulin, cytarabine), the future looks quite promising. Current clinical trials focus mostly on the therapy of cancer, but trials for therapy of pain or Alzheimer’s disease and many others are also underway.

Key words:
marine pharmacology


Sources

1. Mora C., Tittensor D. P., Adl S., Simpson A. G. B., Worm B. How Many Species Are There on Earth and in the Ocean? PLoS Biol. 2011; 9, e1001127.

2. Newton G. G., Abraham E. P. Cephalosporin C, a new antibiotic containing sulphur and D-alpha-aminoadipic acid. Nature 1955; 175, 548.

3. Bo G. Giuseppe Brotzu and the discovery of cephalosporins. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2000; 6 (Suppl 3), 6–9.

4. Radjasa O. K., Vaske Y. M., Navarro G., Vervoort H. C., Tenney K., Linington R. G., Crews P. Highlights of marine invertebrate-derived biosynthetic products: their biomedical potential and possible production by microbial associants. Bioorg. Med. Chem. 2011; 19, 6658–6674.

5. Anjum K., Abbas S. Q., Shah S. A. A., Akhter N., Batool S., Hassan S. S. ul. Marine Sponges as a Drug Treasure. Biomol. Ther. 2016; 24, 347–362.

6. Thomas T. R. A., Kavlekar D. P., LokaBharathi P. A. Marine drugs from sponge-microbe association – a review. Mar. Drugs 2010; 8, 1417–1468.

7. Faulkner D. J., He H. Y., Unson M. D., Bewley C. A., Garson M. J. New metabolites from marine sponges: Are symbionts important? Gazz Chem Ital 1993; 123, 301–307.

8. Haygood M. G., Schmidt E. W., Davidson S. K., Faulkner D. J. Microbial symbionts of marine invertebrates: opportunities for microbial biotechnology. J. Mol. Microbiol. Biotechnol. 1999; 1, 33–43.

9. Vacelet J., Donadey C. Electron microscope study of the association between some sponges and bacteria. J. Exp. Mar. Biol. Ecol. 1977; 301–314.

10. Anand T. P., Bhat A. W., Shouche Y. S., Roy U., Siddharth J., Sarma S. P. Antimicrobial activity of marine bacteria associated with sponges from the waters off the coast of South East India. Microbiol. Res. 2006; 161, 252–262.

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

12. Gerwick W. H., Fenner A. M. Drug Discovery from Marine Microbes. Microb. Ecol. 2013; 65, 800–806.

13. Gomes N. G. M., Dasari R., Chandra S., Kiss R., Kornienko A. Marine Invertebrate Metabolites with Anticancer Activities: Solutions to the “Supply Problem”. Mar. Drugs 2016; 14(5), 98.

14. Schaufelberger D. E., Koleck M. P., Beutler J. A., Vatakis A. M., Alvarado A. B., Andrews P., Marzo L. V., Muschik G. M., Roach J., Ross J. T. The large-scale isolation of bryostatin 1 from Bugula neritina following current good manufacturing practices. J. Nat. Prod. 1991; 54, 1265–1270.

15. Cuevas C., Francesch A. Development of Yondelis (trabectedin, ET-743). A semisynthetic process solves the supply problem. Nat. Prod. Rep. 2009; 26, 322–337.

16. Mendola D. Aquaculture of three phyla of marine invertebrates to yield bioactive metabolites: process developments and economics. Biomol. Eng. 2003; 20, 441–458.

17. Duckworth A. Farming sponges to supply bioactive metabolites and bath sponges: a review. Mar. Biotechnol. N. Y. N 2009; 11, 669–679.

18. Belarbi E. H., Goméz A. C., Chisti Y., Camacho F. C., Grima E. M. Producing drugs from marine sponges. Biotechnol. Adv. 2003; 585–598.

19. Dunlap W. C., Battershill C. N., Liptrot C. H., Cobb R. E., Bourne D. G., Jaspars M., Long P. F., Newman D. J. Biomedicinals from the phytosymbionts of marine invertebrates: a molecular approach. Methods San Diego Calif 2007; 42, 358–376.

20. Prialt SPC. http://www.ema.europa.eu/docs/cs_CZ/document_library/EPAR_-_Product_Information/human/000551/WC500041929.pdf (30. 5. 2017).

21. Bowersox S. S., Luther R. Pharmacotherapeutic potential of omega-conotoxin MVIIA (SNX-111), an N-type neuronal calcium channel blocker found in the venom of Conus magus. Toxicon Off. J. Int. Soc. Toxinology 1998; 36, 1651–1658.

22. Molderings G. J., Likungu J., Göthert M. N-Type calcium channels control sympathetic neurotransmission in human heart atrium. Circulation 2000; 101, 403–407.

23. Hayashi K., Wakino S., Sugano N., Ozawa Y., Homma K., Saruta T. Ca2+ channel subtypes and pharmacology in the kidney. Circ. Res. 2007; 100, 342–353.

24. Gohil K., Bell J. R., Ramachandran J., Miljanich G. P. Neuroanatomical distribution of receptors for a novel voltage-sensitive calcium-channel antagonist, SNX-230 (omega-conopeptide MVIIC). Brain Res. 1994; 653, 258–266.

25. Malmberg A. B., Yaksh T. L. Effect of continuous intrathecal infusion of omega-conopeptides, N-type calcium-channel blockers, on behavior and antinociception in the formalin and hot-plate tests in rats. Pain 1995; 60, 83–90.

26. Webster L. R., Fakata K. L., Charapata S., Fisher R., MineHart M. Open-label, multicenter study of combined intrathecal morphine and ziconotide: addition of morphine in patients receiving ziconotide for severe chronic pain. Pain Med. Malden Mass 2008; 9, 282–290.

27. de la Calle Gil A. B., Peña Vergara I., Cormane Bornacelly M. A., Pajuelo Gallego A. Intrathecal Ziconotide and Morphine for Pain Relief: A Case Series of Eight Patients with Refractory Cancer Pain, Including Five Cases of Neuropathic Pain. Neurol. Ther. 2015; 4, 159–168.

28. Staquet H., Dupoiron D., Nader E., Menei P. Intracerebroventricular Pain Treatment with Analgesic Mixtures including Ziconotide for Intractable Pain. Pain Physician 2016; 19, E905–915.

29. Bowersox S. S., Singh T., Luther R. R. Selective blockade of N-type voltage-sensitive calcium channels protects against brain injury after transient focal cerebral ischemia in rats. Brain Res. 1997; 747, 343–347.

30. Perez-Pinzon M. A., Yenari M. A., Sun G. H., Kunis D. M., Steinberg G. K. SNX-111, a novel, presynaptic N-type calcium channel antagonist, is neuroprotective against focal cerebral ischemia in rabbits. J. Neurol. Sci. 1997; 153, 25–31.

31. Berman R. F., Verweij B. H., Muizelaar J. P. Neurobehavioral protection by the neuronal calcium channel blocker ziconotide in a model of traumatic diffuse brain injury in rats. J. Neurosurg. 2000; 93, 821–828.

32. Rinehart K. L., Holt T. G., Fregeau H. L., Stroh J. G., Keifer P. A., Sun F., Li L. H., Martin D. G. Ecteinascidins 729, 743, 745, 759A, 759B, and 770: potent antitumor agents from the Caribbean tunicate Ecteinascidia turbinata. J. Org. Chem. 1990; 4512–4515.

33. Rath C. M., Janto B., Earl J., Ahmed A., Hu F. Z., Hiller L., Dahlgren M., Kreft R., Yu F., Wolff J. J., Kweon H. K., Christiansen M. A., Håkansson K., Williams R. M., Ehrlich G. D., Sherman D. H. Meta-omic characterization of the marine invertebrate microbial consortium that produces the chemotherapeutic natural product ET-743. ACS Chem. Biol. 2011; 6, 1244–1256.

34. Schofield M. M., Jain S., Porat D., Dick G. J., Sherman D. H. Identification and analysis of the bacterial endosymbiont specialized for production of the chemotherapeutic natural product ET-743. Environ. Microbiol. 2015; 17, 3964–3975.

35. Pommier Y., Kohlhagen G., Bailly C., Waring M., Mazumder A., Kohn K. W. 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 (Mosc.) 1996; 35, 13303–13309.

36. Yondelis SPC. http://www.ema.europa.eu/docs/cs_CZ/document_library/EPAR_-_Product_Information/human/000773/WC500045832.pdf (30. 5. 2017).

37. Dubois E. A., Cohen A. F. Trabectedin. Br. J. Clin. Pharmacol. 2009; 68, 320–321.

38. Takebayashi Y., Pourquier P., Zimonjic D. B., Nakayama K., Emmert S., Ueda T., Urasaki Y., Kanzaki A., Akiyama S. I., Popescu N., Kraemer K. H., Pommier Y. Antiproliferative activity of ecteinascidin 743 is dependent upon transcription-coupled nucleotide-excision repair. Nat. Med. 2001; 7, 961–966.

39. Zewail-Foote M., Li V. S., Kohn H., Bearss D., Guzman M., Hurley L. H. 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. 2001; 8, 1033–1049.

40. Soares D. G., Machado M. S., Rocca C. J., Poindessous V., Ouaret D., Sarasin A., Galmarini C. M., Henriques J. A. P., Escargueil A. E., Larsen A. K. Trabectedin and its C subunit modified analogue PM01183 attenuate nucleotide excision repair and show activity toward platinum-resistant cells. Mol. Cancer Ther. 2011; 10, 1481–1489.

41. Germano G., Frapolli R., Belgiovine C., Anselmo A., Pesce S., Liguori M., Erba E., Uboldi S., Zucchetti M., Pasqualini F., Nebuloni M., van Rooijen N., Mortarini R., Beltrame L., Marchini S., Fuso Nerini I., Sanfilippo R., Casali P. G., Pilotti S., Galmarini C. M., Anichini A., Mantovani A., D’Incalci M., Allavena P. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 2013; 23, 249–262.

42. Bingle L., Brown N. J., Lewis C. E. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J. Pathol. 2002; 196, 254–265.

43. Comito G., Giannoni E., Segura C. P., Barcellos-de-Souza P., Raspollini M. R., Baroni G., Lanciotti M., Serni S., Chiarugi P. Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene 2014; 33, 2423–2431.

44. Komohara Y., Takeya M. CAFs and TAMs: maestros of the tumour microenvironment. J. Pathol. 2017; 241, 313–315.

45. D’Angelo D., Borbone E., Palmieri D., Uboldi S., Esposito F., Frapolli R., Pacelli R., D’Incalci M., Fusco A. The impairment of the High Mobility Group A (HMGA) protein function contributes to the anticancer activity of trabectedin. Eur. J. Cancer Oxf. Engl. 2013; 49, 1142–1151.

46. De Martino M., Forzati F., Arra C., Fusco A., Esposito F. HMGA1-pseudogenes and cancer. Oncotarget 2016; 7, 28724–28735.

47. Kanzaki A., Takebayashi Y., Ren X.-Q., Miyashita H., Mori S., Akiyama S., Pommier Y. Overcoming multidrug drug resistance in P-glycoprotein/MDR1-overexpressing cell lines by ecteinascidin 743. Mol. Cancer Ther. 2002; 1, 1327–1334.

48. Goldstein L. J., Gurtler J., Del Prete S. A., Tjulandin S., Semiglazov V. F., Bayever E., Michiels B. Trabectedin as a single-agent treatment of advanced breast cancer after anthracycline and taxane treatment: a multicenter, randomized, phase II study comparing 2 administration regimens. Clin. Breast Cancer 2014; 14, 396–404.

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

50. Pettit G. R., Kamano Y., Fujii Y., Herald C. L., Inoue M., Brown P., Gust D., Kitahara K., Schmidt J. M., Doubek D. L., Michel C. Marine animal biosynthetic constituents for cancer chemotherapy. J. Nat. Prod. 1981; 44, 482–485.

51. Bai R. L., 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. 1990; 265, 17141–17149.

52. Müller P., Martin K., Theurich S., Schreiner J., Savic S., Terszowski G., Lardinois D., Heinzelmann-Schwarz V. A., Schlaak M., Kvasnicka H-M., Spagnoli G., Dirnhofer S., Speiser D. E., von Bergwelt-Baildon M., Zippelius A. Microtubule-depolymerizing agents used in antibody-drug conjugates induce antitumor immunity by stimulation of dendritic cells. Cancer Immunol. Res. 2014; 2, 741–755.

53. Adcetris SPC. http://www.ema.europa.eu/docs/cs_CZ/document_ library/EPAR_-_Product_Information/human/002455/WC500135055.pdf (30. 5. 2017).

54. Corey K., Cook D., Bekker J., Mugnaini E., Lin J. H. A case of refractory Sézary syndrome with large-cell transformation responsive to brentuximab vedotin. JAMA Dermatol. 2014; 150, 210–212.

55. Mehra T., Ikenberg K., Moos R. M., Benz R., Nair G., Schanz U., Haralambieva E., Hoetzenecker W., Dummer R., French L. E., Guenova E., Cozzio A. Brentuximab as a treatment for CD30+ mycosis fungoides and Sézary syndrome. JAMA Dermatol. 2015; 151, 73–77.

56. Saintes C., Saint-Jean M., Renaut J. J., Dréno B., Quéreux G. Dramatic efficacy of brentuximab vedotin in two patients with epidermotropic cutaneous T-cell lymphomas after treatment failure despite variable CD30 expression. Br. J. Dermatol. 2015; 172, 819–821.

57. Criscuolo M., Fianchi L., Voso M. T., Pagano L. Rapid response of nodular CD30-positive mycosis fungoides to brentuximab vedotin. Br. J. Haematol. 2015; 168, 617.

58. Duvic M., Tetzlaff M. T., Gangar P., Clos A. L., Sui D., Talpur R. Results of a Phase II Trial of Brentuximab Vedotin for CD30+ Cutaneous T-Cell Lymphoma and Lymphomatoid Papulosis. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2015; 33, 3759–3765.

59. Jacobsen E. D., Sharman J. P., Oki Y., Advani R. H., Winter J. N., Bello C. M., Spitzer G., Palanca-Wessels M. C., Kennedy D. A., Levine P., Yang J., Bartlett N. L. Brentuximab vedotin demonstrates objective responses in a phase 2 study of relapsed/refractory DLBCL with variable CD30 expression. Blood 2015; 125, 1394–1402.

60. Holderness B. M., Malhotra S., Levy N. B., Danilov A. V. Brentuximab vedotin demonstrates activity in a patient with plasmablastic lymphoma arising from a background of chronic lymphocytic leukemia. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2013; 31, e197–199.

61. Dabir S., Kresak A., Yang M., Fu P., Wildey G., Dowlati A. CD30 is a potential therapeutic target in malignant mesothelioma. Mol. Cancer Ther. 2015; 14, 740–746.

62. Okouneva T., Azarenko O., Wilson L., Littlefield B. A., Jordan M. A. Inhibition of centromere dynamics by eribulin (E7389) during mitotic metaphase. Mol. Cancer Ther. 2008; 7, 2003–2011.

63. Bai R. L., Paull K. D., Herald C. L., Malspeis L., Pettit G. R., Hamel E. 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. 1991; 266, 15882–15889.

64. Jordan M. A., Kamath K., Manna T., Okouneva T., Miller H. P., Davis C., Littlefield B. A., Wilson L. The primary antimitotic mechanism of action of the synthetic halichondrin E7389 is suppression of microtubule growth. Mol. Cancer Ther. 2005; 4, 1086–1095.

65. Funahashi Y., Okamoto K., Adachi Y., Semba T., Uesugi M., Ozawa Y., Tohyama O., Uehara T., Kimura T., Watanabe H., Asano M., Kawano S., Tizon X., McCracken P. J., Matsui J., Aoshima K., Nomoto K., Oda Y. Eribulin mesylate reduces tumor microenvironment abnormality by vascular remodeling in preclinical human breast cancer models. Cancer Sci. 2014; 105, 1334–1342.

66. Yoshida T., Ozawa Y., Kimura T., Sato Y., Kuznetsov G., Xu S., Uesugi M., Agoulnik S., Taylor N., Funahashi Y., Matsui J. Eribulin mesilate suppresses experimental metastasis of breast cancer cells by reversing phenotype from epithelial-mesenchymal transition (EMT) to mesenchymal-epithelial transition (MET) states. Br. J. Cancer 2014; 110, 1497–1505.

67. Kitahara H., Hirai M., Kato K., Bou-Gharios G., Nakamura H., Kawashiri S. Eribulin sensitizes oral squamous cell carcinoma cells to cetuximab via induction of mesenchymal-to-epithelial transition. Oncol. Rep. 2016.

68. Gavert N., Ben-Ze’ev A. Epithelial-mesenchymal transition and the invasive potential of tumors. Trends Mol. Med. 2008; 14, 199–209.

69. Hirata Y., Uemura D. Halichondrins – antitumor polyether macrolides from a marine sponge. Pure Appl. Chem. 1986; 58.

70. Pettit G. R., Tan R., Gao F., Williams M. D., Doubek D. L., Boyd M. R., Schmidt J. M., Chapuis J. C., Hamel E. Isolation and structure of halistatin 1 from the eastern Indian Ocean marine sponge Phakellia carteri. J. Org. Chem. 1993; 58, 2538–2543.

71. HALAVEN SPC. http://www.ema.europa.eu/docs/cs_CZ/document_library/EPAR_-_Product_Information/human/002084/WC500105112.pdf (30. 5. 2017).

72. Cortes J., O’Shaughnessy J., Loesch D., Blum J. L., Vahdat L. T., Petrakova K., Chollet P., Manikas A., Diéras V., Delozier T., Vladimirov V., Cardoso F., Koh H., Bougnoux P., Dutcus C. E., Seegobin S., Mir D., Meneses N., Wanders J., Twelves C., EMBRACE (Eisai Metastatic Breast Cancer Study Assessing Physician’s Choice Versus E7389) investigators. Eribulin monotherapy versus treatment of physician’s choice in patients with metastatic breast cancer (EMBRACE): a phase 3 open-label randomised study. Lancet Lond. Engl. 2011; 377, 914–923.

73. Schöffski P., Chawla S., Maki R. G., Italiano A., Gelderblom H., Choy E., Grignani G., Camargo V., Bauer S., Rha S. Y., Blay J.-Y., Hohenberger P., D’Adamo D., Guo M., Chmielowski B., Le Cesne A., Demetri G. D., Patel S. R. Eribulin versus dacarbazine in previously treated patients with advanced liposarcoma or leiomyosarcoma: a randomised, open-label, multicentre, phase 3 trial. Lancet Lond. Engl. 2016; 387, 1629–1637.

74. Gitlitz B. J., Tsao-Wei D. D., Groshen S., Davies A., Koczywas M., Belani C. P., Argiris A., Ramalingam S., Vokes E. E., Edelman M., Hoffman P., Ballas M. S., Liu S. V., Gandara D. R. A phase II study of halichondrin B analog eribulin mesylate (E7389) in patients with advanced non-small cell lung cancer previously treated with a taxane: a California cancer consortium trial. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2012; 7, 574–578.

75. Spira A. I., Iannotti N. O., Savin M. A., Neubauer M., Gabrail N. Y., Yanagihara R. H., Zang E. A., Cole P. E., Shuster D., Das A. A phase II study of eribulin mesylate (E7389) in patients with advanced, previously treated non-small-cell lung cancer. Clin. Lung Cancer 2012; 13, 31–38.

76. Mok T. S., Geater S. L., Iannotti N., Thongprasert S., Spira A., Smith D., Lee V., Lim W. T., Reyderman L., Wang B., Gopalakrishna P., Garzon F., Xu L., Reynolds C. Randomized phase II study of two intercalated combinations of eribulin mesylate and erlotinib in patients with previously treated advanced non-small-cell lung cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2014; 25, 1578–1584.

77. de Bono J. S., Molife L. R., Sonpavde G., Maroto J. P., Calvo E., Cartwright T. H., Loesch D. M., Feit K., Das A., Zang E. A., Wanders J., Agoulnik S., Petrylak D. P. Phase II study of eribulin mesylate (E7389) in patients with metastatic castration-resistant prostate cancer stratified by prior taxane therapy. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2012; 23, 1241–1249.

78. Hensley M. L., Kravetz S., Jia X., Iasonos A., Tew W., Pereira L., Sabbatini P., Whalen C., Aghajanian C. A., Zarwan C., Berlin S. Eribulin mesylate (halichondrin B analog E7389) in platinum-resistant and platinum-sensitive ovarian cancer: a 2-cohort, phase 2 study. Cancer 2012; 118, 2403–2410.

79. Schöffski P., Ray-Coquard I. L., Cioffi A., Bui N. B., Bauer S., Hartmann J. T., Krarup-Hansen A., Grünwald V., Sciot R., Dumez H., Blay J-Y., Le Cesne A., Wanders J., Hayward C., Marreaud S., Ouali M., Hohenberger P., European Organisation for Research and Treatment of Cancer (EORTC) Soft Tissue and Bone Sarcoma Group (STBSG). Activity of eribulin mesylate in patients with soft-tissue sarcoma: a phase 2 study in four independent histological subtypes. Lancet Oncol. 2011; 12, 1045–1052.

80. Takezaki Y., Namikawa T., Koyama T., Munekage E., Munekage M., Maeda H., Kitagawa H., Hanazaki K. Antitumor Effects of Eribulin Mesylate in Gemcitabine-resistant Pancreatic Cancer Cell Lines. Anticancer Res. 2016; 36, 6077–6082.

81. Inagaki A., Nakamura T., Wakisaka G. Studies on the mechanism of action of 1-beta-D-arabinofuranosylcytosine as an inhibitor of DNA synthesis in human leukemic leukocytes. Cancer Res. 1969; 29, 2169–2176.

82. Bergmann W., Feeney R. J. Contributions to the study of marine products. XXXII. The nucleosides of sponges. I. J. Org. Chem. 1951; 981–987.

83. CYTARABINE ACCORD SPC. http://www.sukl.cz/modules/medication/download.php?file=SPC76455.pdf&type=spc&as=cytarabine-accord-100-mg-ml-injekcni-infuzni-roztok-spc (30. 5. 2017).

84. CYTOSAR SPC. http://www.sukl.cz/modules/medication/download.php?file=SPC66037.pdf&type=spc&as=cytosar-1-g-spc (30. 5. 2017).

85. ALEXAN SPC. http://www.sukl.cz/modules/medication/download.php?file=SPC50287.pdf&type=spc&as=alexan-50-mg-ml-spc (30. 5. 2017).

86. DEPOCYTE SPC. http://www.ema.europa.eu/docs/cs_CZ/document_library/EPAR_-_Product_Information/human/000317/WC500035649.pdf (30. 5. 2017).

87. Rai K. R., Holland J. F., Glidewell O. J., Weinberg V., Brunner K., Obrecht J. P., Preisler H. D., Nawabi I. W., Prager D., Carey R. W., Cooper M. R., Haurani F., Hutchison J. L., Silver R. T., Falkson G., Wiernik P., Hoagland H. C., Bloomfield C. D., James G. W., Gottlieb A., Ramanan S. V., Blom J., Nissen N. I., Bank A., Ellison R. R., Kung F., Henry P., McIntyre O. R., Kaan S. K. Treatment of acute myelocytic leukemia: a study by cancer and leukemia group B. Blood 1981; 58, 1203–1212.

88. Yates J., Glidewell O., Wiernik P., Cooper M. R., Steinberg D., Dosik H., Levy R., Hoagland C., Henry P., Gottlieb A., Cornell C., Berenberg J., Hutchison J. L., Raich P., Nissen N., Ellison R. R., Frelick R., James G. W., Falkson G., Silver R. T., Haurani F., Green M., Henderson E., Leone L., Holland J. F. Cytosine arabinoside with daunorubicin or adriamycin for therapy of acute myelocytic leukemia: a CALGB study. Blood 1982; 60, 454–462.

89. Perry M. C., Doll D. C., Freter C. E. Chemotherapy source book. 5th ed. Philadelphia: Wolters Kluwer/Lippincott Williams & Wilkins 2012.

90. AML Collaborative Group. A systematic collaborative overview of randomized trials comparing idarubicin with daunorubicin (or other anthracyclines) as induction therapy for acute myeloid leukaemia. AML Collaborative Group. Br. J. Haematol. 1998; 103, 100–109.

91. Li X., Xu S., Tan Y., Chen J. The effects of idarubicin versus other anthracyclines for induction therapy of patients with newly diagnosed leukaemia. Cochrane Database Syst. Rev. 2015; CD010432.

92. Holowiecki J., Grosicki S., Robak T., Kyrcz-Krzemien S., Giebel S., Hellmann A., Skotnicki A., Jedrzejczak W. W., Konopka L., Kuliczkowski K., Zdziarska B., Dmoszynska A., Marianska B., Pluta A., Zawilska K., Komarnicki M., Kloczko J., Sulek K., Haus O., Stella-Holowiecka B., Baran W., Jakubas B., Paluszewska M., Wierzbowska A., Kielbinski M., Jagoda K., Polish Adult Leukemia Group (PALG). Addition of cladribine to daunorubicin and cytarabine increases complete remission rate after a single course of induction treatment in acute myeloid leukemia. Multicenter, phase III study. Leukemia 2004; 18, 989–997.

93. Holowiecki J., Grosicki S., Giebel S., Robak T., Kyrcz-Krzemien S., Kuliczkowski K., Skotnicki A. B., Hellmann A., Sulek K., Dmoszynska A., Kloczko J., Jedrzejczak W. W., Zdziarska B., Warzocha K., Zawilska K., Komarnicki M., Kielbinski M., Piatkowska-Jakubas B., Wierzbowska A., Wach M., Haus O. Cladribine, but not fludarabine, added to daunorubicin and cytarabine during induction prolongs survival of patients with acute myeloid leukemia: a multicenter, randomized phase III study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2012; 30, 2441–2448.

94. Wiedower E., Jamy O., Martin M. G. Induction of Acute Myeloid Leukemia with Idarubicin, Cytarabine and Cladribine. Anticancer Res. 2015; 35, 6287–6290.

95. Fridle C., Medinger M., Wilk M. C., Seipel K., Passweg J., Manz M. G., Pabst T. Cladribine, cytarabine and idarubicin (CLA-Ida) salvage chemotherapy in relapsed acute myeloid leukemia (AML). Leuk. Lymphoma 2016; 1–8.

96. Caballero M. D., Rubio V., Rifon J., Heras I., García-Sanz R., Vázquez L., Vidriales B., del Cañizo M. C., Corral M., Gonzalez M., León A., Jean-Paul E., Rocha E., Moraleda J. M., San Miguel J. F. BEAM chemotherapy followed by autologous stem cell support in lymphoma patients: analysis of efficacy, toxicity and prognostic factors. Bone Marrow Transplant. 1997; 20, 451–458.

97. Sharma A., Kayal S., Iqbal S., Malik P. S., Raina V. Comparison of BEAM vs. LEAM regimen in autologous transplant for lymphoma at AIIMS. SpringerPlus 2013; 2, 489.

98. Kim J.-W., Lee H. J., Yi H. G., Kim B.-S., Bang S.-M., Kim J. S., Kim I., Yoon S.-S., Lee J. S., Kim C. S., Park S., Kim B. K. Mitoxantrone, etoposide, cytarabine, and melphalan (NEAM) followed by autologous stem cell transplantation for patients with chemosensitive aggressive non-Hodgkin lymphoma. Am. J. Hematol. 2012; 87, 479–483.

99. Probíhající studie s cytarabinem v EU. https://www.clinicaltrialsregister.eu/ctr-search/search?query=Cytarabine+&status=ongoing (30. 5. 2017).

100. Probíhající studie s cytarabinem v USA. https://www.clinicaltrials.gov/ct2/results?term=cytarabine&recr=Open&no_unk=Y&show_flds=Y (30. 5. 2017).

101. Sagar S., Kaur M., Minneman K. P. Antiviral lead compounds from marine sponges. Mar. Drugs 2010; 8, 2619–2638.

102. Privat de Garilhe M., De Rudder J. Effect of 2 arabinose nucleosides on the multiplication of herpes virus and vaccine in cell culture. Comptes Rendus Hebd. Seances Acad. Sci. 1964; 259, 2725–2728.

103. Cozzarelli N. R. The mechanism of action of inhibitors of DNA synthesis. Annu. Rev. Biochem. 1977; 46, 641–668.

104. Iwatsubo K., Bravo C., Uechi M., Baljinnyam E., Nakamura T., Umemura M., Lai L., Gao S., Yan L., Zhao X., Park M., Qiu H., Okumura S., Iwatsubo M., Vatner D. E., Vatner S. F., Ishikawa Y. Prevention of heart failure in mice by an antiviral agent that inhibits type 5 cardiac adenylyl cyclase. Am. J. Physiol. Heart Circ. Physiol. 2012; 302, H2622–2628.

105. Vatner S. F., Pachon R. E., Vatner D. E. Inhibition of adenylyl cyclase type 5 increases longevity and healthful aging through oxidative stress protection. Oxid. Med. Cell. Longev. 2015; 2015, 250310.

106. Bravo C. A., Vatner D. E., Pachon R., Zhang J., Vatner S. F. A Food and Drug Administration-Approved Antiviral Agent that Inhibits Adenylyl Cyclase Type 5 Protects the Ischemic Heart Even When Administered after Reperfusion. J. Pharmacol. Exp. Ther. 2016; 357, 331–336.

107. Nakamura T., Fujita T., Kishimura M., Suita K., Hidaka Y., Cai W., Umemura M., Yokoyama U., Uechi M., Ishikawa Y. Vidarabine, an Anti-Herpes Virus Agent, Protects Against the Development of Heart Failure With Relatively Mild Side-Effects on Cardiac Function in a Canine Model of Pacing-Induced Dilated Cardiomyopathy. Circ. J. Off. J. Jpn. Circ. Soc. 2016; 80, 2496–2505.

108. Leal J. F. M., Martínez-Díez M., García-Hernández V., Moneo V., Domingo A., Bueren-Calabuig J. A., Negri A., Gago F., Guillén-Navarro M. J., Avilés P., Cuevas C., García-Fernández L. F., Galmarini C. M. PM01183, a new DNA minor groove covalent binder with potent in vitro and in vivo anti-tumour activity. Br. J. Pharmacol. 2010; 161, 1099–1110.

109. Santamaría Nuñez G., Robles C. M. G., Giraudon C., Martínez-Leal J. F., Compe E., Coin F., Aviles P., Galmarini C. M., Egly J.-M. Lurbinectedin specifically triggers the degradation of phosphorylated rna polymerase ii and the formation of DNA breaks in cancer cells. Mol. Cancer Ther. 2016; 15, 2399–2412.

110. Céspedes M. V., Guillén M. J., López-Casas P. P., Sarno F., Gallardo A., Álamo P., Cuevas C., Hidalgo M., Galmarini C. M., Allavena P., Avilés P., Mangues R. Lurbinectedin induces depletion of tumor-associated macrophages (TAM), an essential component of its in vivo synergism with gemcitabine. Dis. Model. Mech. 2016.

111. Vidal A., Muñoz C., Guillén M-J., Moretó J., Puertas S., Martínez-Iniesta M., Figueras A., Padullés L., García-Rodriguez F. J., Berdiel-Acer M., Pujana M. A., Salazar R., Gil-Martin M., Martí L., Ponce J., Molleví D. G., Capella G., Condom E., Viñals F., Huertas D., Cuevas C., Esteller M., Avilés P., Villanueva A. Lurbinectedin (PM01183), a new DNA minor groove binder, inhibits growth of orthotopic primary graft of cisplatin-resistant epithelial ovarian cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2012; 18, 5399–5411.

112. Elez M. E., Tabernero J., Geary D., Macarulla T., Kang S. P., Kahatt C., Pita A. S-M., Teruel C. F., Siguero M., Cullell-Young M., Szyldergemajn S., Ratain M. J. First-in-human phase I study of Lurbinectedin (PM01183) in patients with advanced solid tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014; 20, 2205–2214.

113. CORAIL. Clinical Trial of Lurbinectedin (PM01183) in Platinum Resistant Ovarian Cancer Patients (CORAIL). https://clinicaltrials.gov/ct2/show/NCT02421588 (30. 5. 2017).

114. NCT02448537. A Phase II Multi-Strata Study of PM01183 as a Single Agent or in Combination With Conventional Chemotherapy in Metastatic and/or Unresectable Sarcomas. https://clinicaltrials.gov/ct2/show/NCT02448537 (30. 5. 2017).

115. NCT01525589. A Phase II Clinical Trial of PM01183 in BRCA 1/2-Associated or Unselected Metastatic Breast Cancer. https://clinicaltrials.gov/ct2/show/NCT01525589 (30. 5. 2017).

116. Lago J., Rodríguez L. P., Blanco L., Vieites J. M., Cabado A. G. Tetrodotoxin, an Extremely Potent Marine Neurotoxin: Distribution, Toxicity, Origin and Therapeutical Uses. Mar. Drugs 2015; 13, 6384–6406.

117. Narahashi T. Tetrodotoxin – A brief history. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2008; 84, 147–154.

118. Mosher H. S., Fuhrman F. A., Buchwald H. D., Fischer H. G. Tarichatoxin-tetrodotoxin: a potent neurotoxin. Science 1964; 144, 1100–1110.

119. Kim Y. H., Brown G. B., Mosher F. A. Tetrodotoxin: Occurrence in atelopid frogs of Costa Rica. Science 1975; 189, 151–152.

120. Sheumack D. D., Howden M. E., Spence I., Quinn R. J. Maculotoxin: a neurotoxin from the venom glands of the octopus Hapalochlaena maculosa identified as tetrodotoxin. Science 1978; 199, 188–189.

121. Noguchi T., Jeon J. K., Arakawa O., Sugita H., Deguchi Y., Shida Y., Hashimoto K. Occurrence of tetrodotoxin and anhydrotetrodotoxin in Vibrio sp. isolated from the intestines of a xanthid crab, Atergatis floridus. J. Biochem. (Tokyo) 1986; 99, 311–314.

122. Chau R., Kalaitzis J. A., Neilan B. A. On the origins and biosynthesis of tetrodotoxin. Aquat. Toxicol. Amst. Neth. 2011; 104, 61–72.

123. Yu V. C.-H., Yu P. H.-F., Ho K.-C., Lee F. W.-F. Isolation and Identification of a New Tetrodotoxin-Producing Bacterial Species, Raoultella terrigena, from Hong Kong Marine Puffer Fish Takifugu niphobles. Mar. Drugs 2011; 9, 2384–2396.

124. Chau J., Ciufolini M. A. The Chemical Synthesis of Tetrodoxin: An Ongoing Quest. Mar. Drugs 2011; 9, 2046–2074.

125. Shi J., Liu T.-T., Wang X., Epstein D. H., Zhao L.-Y., Zhang X.-L., Lu L. Tetrodotoxin reduces cue-induced drug craving and anxiety in abstinent heroin addicts. Pharmacol. Biochem. Behav. 2009; 92, 603–607.

126. Song H., Li J., Lu C.-L., Kang L., Xie L., Zhang Y.-Y., Zhou X.-B., Zhong S. Tetrodotoxin alleviates acute heroin withdrawal syndrome: a multicentre, randomized, double-blind, placebo-controlled study. Clin. Exp. Pharmacol. Physiol. 2011; 38, 510–514.

127. Kostyuk P. G., Veselovsky N. S., Tsyndrenko A. Y. Ionic currents in the somatic membrane of rat dorsal root ganglion neurons-I. Sodium currents. Neuroscience 1981; 6, 2423–2430.

128. Luiz A. P., Wood J. N. Sodium Channels in Pain and Cancer: New Therapeutic Opportunities. Adv. Pharmacol. San Diego Calif 2016; 75, 153–178.

129. McEntire D. M., Kirkpatrick D. R., Dueck N. P., Kerfeld M. J., Smith T. A., Nelson T. J., Reisbig M. D., Agrawal D. K. Pain transduction: a pharmacologic perspective. Expert Rev. Clin. Pharmacol. 2016; 9, 1069–1080.

130. Akopian A. N., Souslova V., England S., Okuse K., Ogata N., Ure J., Smith A., Kerr B. J., McMahon S. B., Boyce S., Hill R., Stanfa L. C., Dickenson A. H., Wood J. N. The tetrodotoxin-resistant sodium channel SNS has a specialized function in pain pathways. Nat. Neurosci. 1999; 2, 541–548.

131. Ogata N., Ohishi Y. Molecular diversity of structure and function of the voltage-gated Na+ channels. Jpn. J. Pharmacol. 2002; 88, 365–377.

132. Strassman A. M., Raymond S. A. Electrophysiological evidence for tetrodotoxin-resistant sodium channels in slowly conducting dural sensory fibers. J. Neurophysiol. 1999; 81, 413–424.

133. Pinto V., Derkach V. A., Safronov B. V. Role of TTX-sensitive and TTX-resistant sodium channels in Adelta- and C-fiber conduction and synaptic transmission. J. Neurophysiol. 2008; 99, 617–628.

134. Blair N. T., Bean B. P. Roles of tetrodotoxin (TTX)-sensitive Na+ current, TTX-resistant Na+ current, and Ca2+ current in the action potentials of nociceptive sensory neurons. J. Neurosci. Off. J. Soc. Neurosci. 2002; 22, 10277–10290.

135. Zimmermann K., Leffler A., Babes A., Cendan C. M., Carr R. W., Kobayashi J., Nau C., Wood J. N., Reeh P. W. Sensory neuron sodium channel Nav1.8 is essential for pain at low temperatures. Nature 2007; 447, 855–858.

136. TEC-006. Safety & Efficacy Study of Subcutaneous Tetrodotoxin for Moderate to Severe Inadequately Controlled Cancer-related Pain (TEC-006). https://clinicaltrials.gov/ct2/show/NCT00725114 (30. 5. 2017).

137. TTX-CINP-201. The Purpose of This Study is to Determine if Tetrodotoxin (TTX) is Effective in the Treatment of Pain Resulting From Chemotherapy Treatment (TTX-CINP-201). https://clinicaltrials.gov/ct2/show/NCT01655823 (30. 5. 2017).

138. Ott P. A., Hamid O., Pavlick A. C., Kluger H., Kim K. B., Boasberg P. D., Simantov R., Crowley E., Green J. A., Hawthorne T., Davis T. A., Sznol M., Hwu P. Phase I/II Study of the Antibody-Drug Conjugate Glembatumumab Vedotin in Patients With Advanced Melanoma. J. Clin. Oncol. 2014; 32, 3659–3666.

139. Weterman M. A., Ajubi N., van Dinter I. M., Degen W. G., van Muijen G. N., Ruitter D. J., Bloemers H. P. nmb, a novel gene, is expressed in low-metastatic human melanoma cell lines and xenografts. Int. J. Cancer 1995; 60, 73–81.

140. Tse K. F., Jeffers M., Pollack V. A., McCabe D. A., Shadish M. L., Khramtsov N. V., Hackett C. S., Shenoy S. G., Kuang B., Boldog F. L., MacDougall J. R., Rastelli L., Herrmann J., Gallo M., Gazit-Bornstein G., Senter P. D., Meyer D. L., Lichenstein H. S., LaRochelle W. J. CR011, a fully human monoclonal antibody-auristatin E conjugate, for the treatment of melanoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006; 12, 1373–1382.

141. Kuan C.-T., Wakiya K., Dowell J. M., Herndon J. E., Reardon D. A., Graner M. W., Riggins G. J., Wikstrand C. J., Bigner D. D. Glycoprotein nonmetastatic melanoma protein B, a potential molecular therapeutic target in patients with glioblastoma multiforme. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006; 12, 1970–1982.

142. Rose A. A. N., Grosset A-A., Dong Z., Russo C., Macdonald P. A., Bertos N. R., St-Pierre Y., Simantov R., Hallett M., Park M., Gaboury L., Siegel P. M. Glycoprotein nonmetastatic B is an independent prognostic indicator of recurrence and a novel therapeutic target in breast cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2010; 16, 2147–2156.

143. Roth M., Barris D. M., Piperdi S., Kuo V., Everts S., Geller D., Houghton P., Kolb E. A., Hawthorne T., Gill J., Gorlick R. Targeting Glycoprotein NMB With Antibody-Drug Conjugate, Glembatumumab Vedotin, for the Treatment of Osteosarcoma. Pediatr. Blood Cancer 2016; 63, 32–38.

144. Oyewumi M. O., Manickavasagam D., Novak K., Wehrung D., Paulic N., Moussa F. M., Sondag G. R., Safadi F. F. Osteoactivin (GPNMB) ectodomain protein promotes growth and invasive behavior of human lung cancer cells. Oncotarget 2016; 7, 13932–13944.

145. Bendell J., Saleh M., Rose A. A. N., Siegel P. M., Hart L., Sirpal S., Jones S., Green J., Crowley E., Simantov R., Keler T., Davis T., Vahdat L. Phase I/II study of the antibody-drug conjugate glembatumumab vedotin in patients with locally advanced or metastatic breast cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2014; 32, 3619–3625.

146. Yardley D. A., Weaver R., Melisko M. E., Saleh M. N., Arena F. P., Forero A., Cigler T., Stopeck A., Citrin D., Oliff I., Bechhold R., Loutfi R., Garcia A. A., Cruickshank S., Crowley E., Green J., Hawthorne T., Yellin M. J., Davis T. A., Vahdat L. T. EMERGE: A randomized phase ii study of the antibody-drug conjugate glembatumumab vedotin in advanced glycoprotein nmb-expressing breast cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2015; 33, 1609–1619.

147. NCT01997333. Study of Glembatumumab Vedotin (CDX-011) in Patients With Metastatic, gpNMB Over-Expressing, Triple Negative Breast Cancer (METRIC). https://clinicaltrials.gov/ct2/show/NCT01997333 (30. 5. 2017).

148. Kitagaki J., Shi G., Miyauchi S., Murakami S., Yang Y. Cyclic depsipeptides as potential cancer therapeutics. Anticancer. Drugs 2015; 26, 259–271.

149. Rinehart K. L., Kishore V., Bible K. C., Sakai R., Sullins D. W., Li K. M. Didemnins and tunichlorin: novel natural products from the marine tunicate Trididemnum solidum. J. Nat. Prod. 1988; 51, 1–21.

150. Crampton S. L., Adams E. G., Kuentzel S. L., Li L. H., Badiner G., Bhuyan B. K. Biochemical and cellular effects of didemnins A and B. Cancer Res. 1984; 44, 1796–1801.

151. Jiang T. L., Liu R. H., Salmon S. E. Antitumor activity of didemnin B in the human tumor stem cell assay. Cancer Chemother. Pharmacol. 1983; 11, 1–4.

152. Motzer R., Scher H., Bajorin D., Sternberg C., Bosl G. J. Phase II trial of Didemnin B in patients with advanced renal cell carcinoma. Invest. New Drugs 1990; 8, 391–392.

153. Shin D. M., Holoye P. Y., Murphy W. K., Forman A., Papasozomenos S. C., Hong W. K., Raber M. Phase I/II clinical trial of didemnin B in non-small-cell lung cancer: neuromuscular toxicity is dose-limiting. Cancer Chemother. Pharmacol. 1991; 29, 145–149.

154. Cain J. M., Liu P. Y., Alberts D. E., Gallion H. H., Laufman L., O’Sullivan J., Weiss G., Bickers J. N. Phase II trial of didemnin-B in advanced epithelial ovarian cancer. A Southwest Oncology Group study. Invest. New Drugs 1992; 10, 23–24.

155. Jacobs A. J., Blessing J. A., Munoz A. A phase II trial of didemnin B (NSC No. 325319) in advanced and recurrent cervical carcinoma: a Gynecologic Oncology Group study. Gynecol. Oncol. 1992; 44, 268–270.

156. Sakai R., Rinehart K. L., Kishore V., Kundu B., Faircloth G., Gloer J. B., Carney J. R., Namikoshi M., Sun F., Hughes R. G., García Grávalos D., de Quesada T. G., Wilson G. R., Heid R. M. Structure-activity relationships of the didemnins. J. Med. Chem. 1996; 39, 2819–2834.

157. Broggini M., Marchini S. V., Galliera E., Borsotti P., Taraboletti G., Erba E., Sironi M., Jimeno J., Faircloth G. T., Giavazzi R., D’Incalci M. 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 2003; 17, 52–59.

158. Cuadrado A., Garcia-Fernandez L. F., Gonzalez L., Suarez Y., Losada A., Alcaide V., Martinez T., Fernandez-Sousa J. M., Sanchez-Puelles J. M., Munoz A. 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. 2003; 278, 241–250.

159. González-Santiago L., Suárez Y., Zarich N., Muñoz-Alonso M. J., Cuadrado A., Martínez T., Goya L., Iradi A., Sáez-Tormo G., Maier J. V., Moorthy A., Cato A. C. B., Rojas J. M., Muñoz A. Aplidin induces JNK-dependent apoptosis in human breast cancer cells via alteration of glutathione homeostasis, Rac1 GTPase activation, and MKP-1 phosphatase downregulation. Cell Death Differ. 2006; 13, 1968–1981.

160. Bravo S. B., García-Rendueles M. E. R., Seoane R., Dosil V., Cameselle-Teijeiro J., López-Lázaro L., Zalvide J., Barreiro F., Pombo C. M., Alvarez C. V. Plitidepsin has a cytostatic effect in human undifferentiated (anaplastic) thyroid carcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2005; 11, 7664–7673.

161. Biscardi M., Caporale R., Balestri F., Gavazzi S., Jimeno J., Grossi A. VEGF inhibition and cytotoxic effect of aplidin in leukemia cell lines and cells from acute myeloid leukemia. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2005; 16, 1667–1674.

162. Taraboletti G., Poli M., Dossi R., Manenti L., Borsotti P., Faircloth G. T., Broggini M., D’Incalci M., Ribatti D., Giavazzi R. Antiangiogenic activity of aplidine, a new agent of marine origin. Br. J. Cancer 2004; 90, 2418–2424.

163. Plummer R., Lorigan P., Brown E., Zaucha R., Moiseyenko V., Demidov L., Soriano V., Chmielowska E., Andrés R., Kudryavtseva G., Kahatt C., Szyldergemajn S., Extremera S., de Miguel B., Cullell-Young M., Calvert H. Phase I-II study of plitidepsin and dacarbazine as first-line therapy for advanced melanoma. Br. J. Cancer 2013; 109, 1451–1459.

164. Mateos M. V., Cibeira M. T., Richardson P. G., Prosper F., Oriol A., de la Rubia J., Lahuerta J. J., García-Sanz R., Extremera S., Szyldergemajn S., Corrado C., Singer H., Mitsiades C. S., Anderson K. C., Bladé J., San Miguel J. Phase II clinical and pharmacokinetic study of plitidepsin 3-hour infusion every two weeks alone or with dexamethasone in relapsed and refractory multiple myeloma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2010; 16, 3260–3269.

165. Schöffski P., Guillem V., Garcia M., Rivera F., Tabernero J., Cullell M., Lopez-Martin J. A., Pollard P., Dumez H., del Muro X. G., Paz-Ares L. Phase II randomized study of Plitidepsin (Aplidin), alone or in association with L-carnitine, in patients with unresectable advanced renal cell carcinoma. Mar. Drugs 2009; 7, 57–70.

166. Pardanani A., Tefferi A., Guglielmelli P., Bogani C., Bartalucci N., Rodríguez J., Extremera S., Pérez I., Alfaro V., Vannucchi A. M. Evaluation of plitidepsin in patients with primary myelofibrosis and post polycythemia vera/essential thrombocythemia myelofibrosis: results of preclinical studies and a phase II clinical trial. Blood Cancer J. 2015; 5, e286.

167. Dumez H., Gallardo E., Culine S., Galceran J. C., Schöffski P., Droz J. P., Extremera S., Szyldergemajn S., Fléchon A. Phase II study of biweekly plitidepsin as second-line therapy for advanced or metastatic transitional cell carcinoma of the urothelium. Mar. Drugs 2009; 7, 451–463.

168. Eisen T., Thatcher N., Leyvraz S., Miller W. H., Couture F., Lorigan P., Lüthi F., Small D., Tanovic A., O’Brien M. Phase II study of weekly plitidepsin as second-line therapy for small cell lung cancer. Lung Cancer Amst. Neth. 2009; 64, 60–65.

169. Hamann M. T., Otto C. S., Scheuer P. J., Dunbar D. C. Kahalalides: Bioactive Peptides from a Marine Mollusk Elysia rufescens and Its Algal Diet Bryopsis sp.(1). J. Org. Chem. 1996; 61, 6594–6600.

170. Suárez Y., González L., Cuadrado A., Berciano M., Lafarga M., Muñoz A. Kahalalide F, a new marine-derived compound, induces oncosis in human prostate and breast cancer cells. Mol. Cancer Ther. 2003; 2, 863–872.

171. 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. Pharmacol. 2005; 68, 502–510.

172. Martín-Algarra S., Espinosa E., Rubió J., López López J. J., Manzano J. L., Carrión L. A., Plazaola A., Tanovic A., Paz-Ares L. Phase II study of weekly Kahalalide F in patients with advanced malignant melanoma. Eur. J. Cancer Oxf. Engl. 2009; 1990 45, 732–735.

173. Serova M., de Gramont A., Bieche I., Riveiro M. E., Galmarini C. M., Aracil M., Jimeno J., Faivre S., Raymond E. Predictive Factors of Sensitivity to Elisidepsin, a Novel Kahalalide F-Derived Marine Compound. Mar. Drugs 2013; 11, 944–959.

174. Petty R., Anthoney A., Metges J-P., Alsina M., Gonçalves A., Brown J., Montagut C., Gunzer K., Laus G., Iglesias Dios J. L., Miguel-Lillo B., Bohan P., Salazar R. Phase Ib/II study of elisidepsin in metastatic or advanced gastroesophageal cancer (IMAGE trial). Cancer Chemother. Pharmacol. 2016; 77, 819–827.

175. Oku N., Gustafson K. R., Cartner L. K., Wilson J. A., Shigematsu N., Hess S., Pannell L. K., Boyd M. R., McMahon J. B. Neamphamide A, a new HIV-inhibitory depsipeptide from the Papua New Guinea marine sponge Neamphius huxleyi. J. Nat. Prod. 2004; 67, 1407–1411.

176. Yamano Y., Arai M., Kobayashi M. Neamphamide B, new cyclic depsipeptide, as an anti-dormant mycobacterial substance from a Japanese marine sponge of Neamphius sp. Bioorg. Med. Chem. Lett. 2012; 22, 4877–4881.

177. Moore K. S., Wehrli S., Roder H., Rogers M., Forrest J. N., McCrimmon D., Zasloff M. Squalamine: an aminosterol antibiotic from the shark. Proc. Natl. Acad. Sci. U. S. A. 1993; 90, 1354–1358.

178. Sills A. K., Williams J. I., Tyler B. M., Epstein D. S., Sipos E. P., Davis J. D., McLane M. P., Pitchford S., Cheshire K., Gannon F. H., Kinney W. A., Chao T. L., Donowitz M., Laterra J., Zasloff M., Brem H. Squalamine inhibits angiogenesis and solid tumor growth in vivo and perturbs embryonic vasculature. Cancer Res. 1998; 58, 2784–2792.

179. Bhargava P., Marshall J. L., Dahut W., Rizvi N., Trocky N., Williams J. I., Hait H., Song S., Holroyd K. J., Hawkins M. J. A phase I and pharmacokinetic study of squalamine, a novel antiangiogenic agent, in patients with advanced cancers. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2001; 7, 3912–3919.

180. Herbst R. S., Hammond L. A., Carbone D. P., Tran H. T., Holroyd K. J., Desai A., Williams J. I., Bekele B. N., Hait H., Allgood V., Solomon S., Schiller J. H. A phase I/IIA trial of continuous five-day infusion of squalamine lactate (MSI-1256F) plus carboplatin and paclitaxel in patients with advanced non-small cell lung cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2003; 9, 4108–4115.

181. NCT01769183. Squalamine for the Treatment in Proliferative Diabetic Retinopathy. https://clinicaltrials.gov/ct2/show/NCT01769183 (30. 5. 2017).

182. NCT01678963. Efficacy and Safety of Squalamine Lactate Eye Drops in Subjects With Neovascular (Wet) Age-related Macular Degeneration (AMD). https://clinicaltrials.gov/ct2/show/NCT01678963 (30. 5. 2017).

183. Kem W. R. A study of the occurrence of anabaseine in Paranemertes and other nemertines. Toxicon Off. J. Int. Soc. Toxinology 1971; 9, 23–32.

184. Wheeler J. W., Olubajo O., Storm C. B., Duffield R. M. Anabaseine: venom alkaloid of aphaenogaster ants. Science 1981; 211, 1051–1052.

185. Kem W., Soti F., Wildeboer K., LeFrancois S., MacDougall K., Wei D.-Q., Chou K.-C., Arias H. R. The Nemertine Toxin Anabaseine and Its Derivative DMXBA (GTS-21): Chemical and Pharmacological Properties. Mar. Drugs 2006; 4, 255–273.

186. Berg D. K., Conroy W. G. Nicotinic alpha 7 receptors: synaptic options and downstream signaling in neurons. J. Neurobiol. 2002; 53, 512–523.

187. Wonnacott S., Barik J., Dickinson J., Jones I. W. Nicotinic receptors modulate transmitter cross talk in the CNS: nicotinic modulation of transmitters. J. Mol. Neurosci. MN 2006; 30, 137–140.

188. Hajós M., Rogers B. N. Targeting alpha7 nicotinic acetylcholine receptors in the treatment of schizophrenia. Curr. Pharm. Des. 2010; 16, 538–554.

189. Deutsch S. I., Schwartz B. L., Schooler N. R., Brown C. H., Rosse R. B., Rosse S. M. Targeting alpha-7 nicotinic neurotransmission in schizophrenia: a novel agonist strategy. Schizophr. Res. 2013; 148, 138–144.

190. Beinat C., Banister S. D., Herrera M., Law V., Kassiou M. The therapeutic potential of α7 nicotinic acetylcholine receptor (α7 nAChR) agonists for the treatment of the cognitive deficits associated with schizophrenia. CNS Drugs 2015; 29, 529–542.

191. Hashimoto K. Targeting of α7 Nicotinic Acetylcholine Receptors in the Treatment of Schizophrenia and the Use of Auditory Sensory Gating as a Translational Biomarker. Curr. Pharm. Des. 2015; 21, 3797–3806.

192. Kem W. R. The brain alpha7 nicotinic receptor may be an important therapeutic target for the treatment of Alzheimer’s disease: studies with DMXBA (GTS-21). Behav. Brain Res. 2000; 113, 169–181.

193. Vallés A. S., Borroni M. V., Barrantes F. J. Targeting brain α7 nicotinic acetylcholine receptors in Alzheimer’s disease: rationale and current status. CNS Drugs 2014; 28, 975–987.

194. Wessler I., Kirkpatrick C. J. Acetylcholine beyond neurons: the non-neuronal cholinergic system in humans. Br. J. Pharmacol. 2008; 154, 1558–1571.

195. Reardon C. Neuro-immune interactions in the cholinergic anti-inflammatory reflex. Immunol. Lett. 2016; 178, 92–96.

196. Yue Y., Liu R., Cheng W., Hu Y., Li J., Pan X., Peng J., Zhang P. GTS-21 attenuates lipopolysaccharide-induced inflammatory cytokine production in vitro by modulating the Akt and NF-κB signaling pathway through the α7 nicotinic acetylcholine receptor. Int. Immunopharmacol. 2015; 29, 504–512.

197. Giebelen I. A. J., van Westerloo D. J., LaRosa G. J., de Vos A. F., van der Poll T. Stimulation of alpha 7 cholinergic receptors inhibits lipopolysaccharide-induced neutrophil recruitment by a tumor necrosis factor alpha-independent mechanism. Shock Augusta Ga 2007; 27, 443–447.

198. Kox M., Pompe J. C., Gordinou de Gouberville M. C., van der Hoeven J. G., Hoedemaekers C. W., Pickkers P. Effects of the α7 nicotinic acetylcholine receptor agonist GTS-21 on the innate immune response in humans. Shock Augusta Ga 2011; 36, 5–11.

199. Kihara T., Shimohama S., Sawada H., Kimura J., Kume T., Kochiyama H., Maeda T., Akaike A. Nicotinic receptor stimulation protects neurons against beta-amyloid toxicity. Ann. Neurol. 1997; 42, 159–163.

200. Jin Y., Tsuchiya A., Kanno T., Nishizaki T. Amyloid-β peptide increases cell surface localization of α7 ACh receptor to protect neurons from amyloid β-induced damage. Biochem. Biophys. Res. Commun. 2015; 468, 157–160.

201. Lombardo S., Maskos U. Role of the nicotinic acetylcholine receptor in Alzheimer’s disease pathology and treatment. Neuropharmacology 2015; 96, 255–262.

202. Kitagawa H., Takenouchi T., Azuma R., Wesnes K. A., Kramer W. G., Clody D. E., Burnett A. L. Safety, pharmacokinetics, and effects on cognitive function of multiple doses of GTS-21 in healthy, male volunteers. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2003; 28, 542–551.

203. Olincy A., Harris J. G., Johnson L. L., Pender V., Kongs S., Allensworth D., Ellis J., Zerbe G. O., Leonard S., Stevens K. E., Stevens J. O., Martin L., Adler L. E., Soti F., Kem W. R., Freedman R. Proof-of-concept trial of an alpha7 nicotinic agonist in schizophrenia. Arch. Gen. Psychiatry 2006; 63, 630–638.

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

205. NCT00414622. GTS21-201 for Alzheimer Disease:GTS-21 Administered Daily for 28 Days to Participants With Probable Alzheimer’s Disease.https://clinicaltrials.gov/ct2/show/NCT00414622 (30. 5. 2017).

206. NCT00419445. Safety and Efficacy of GTS21 in Adults With Attention-deficit Hyperactivity Disorder. https://clinicaltrials.gov/ct2/show/NCT00419445 (30. 5. 2017).

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

208. Davidson S. K., Allen S. W., Lim G. E., Anderson C. M., Haygood M. G. Evidence for the biosynthesis of bryostatins by the bacterial symbiont ‘Candidatus Endobugula sertula’ of the bryozoan Bugula neritina. Appl. Environ. Microbiol. 2001; 67, 4531–4537.

209. Sudek S., Lopanik N. B., Waggoner L. E., Hildebrand M., Anderson C., Liu H., Patel A., Sherman D. H., Haygood M. G. Identification of the putative bryostatin polyketide synthase gene cluster from ‘Candidatus Endobugula sertula’, the uncultivated microbial symbiont of the marine bryozoan Bugula neritina. J. Nat. Prod. 2007; 70, 67–74.

210. Kollár P., Rajchard J., Balounová Z., Pazourek J. Marine natural products: bryostatins in preclinical and clinical studies. Pharm. Biol. 2014; 52, 237–242.

211. Russo P., Kisialiou A., Lamonaca P., Moroni R., Prinzi G., Fini M. New Drugs from Marine Organisms in Alzheimer’s Disease. Mar. Drugs 2015; 14, 5.

212. Stone R. M., Sariban E., Pettit G. R., Kufe D. W. Bryostatin 1 activates protein kinase C and induces monocytic differentiation of HL-60 cells. Blood 1988; 72, 208–213.

213. Nelson T. J., Alkon D. L. Neuroprotective versus tumorigenic protein kinase C activators. Trends Biochem. Sci. 2009; 34, 136–145.

214. Black J. D. Protein kinase C-mediated regulation of the cell cycle. Front. Biosci. J. Virtual Libr. 2000; 5, D406–423.

215. Zhang X., Zhang R., Zhao H., Cai H., Gush K. A., Kerr R. G., Pettit G. R., Kraft A. S. Preclinical pharmacology of the natural product anticancer agent bryostatin 1, an activator of protein kinase C. Cancer Res. 1996; 56, 802–808.

216. Asiedu C., Biggs J., Lilly M., Kraft A. S. Inhibition of leukemic cell growth by the protein kinase C activator bryostatin 1 correlates with the dephosphorylation of cyclin-dependent kinase 2. Cancer Res. 1995; 55, 3716–3720.

217. Philip P. A., Rea D., Thavasu P., Carmichael J., Stuart N. S., Rockett H., Talbot D. C., Ganesan T., Pettit G. R., Balkwill F. Phase I study of bryostatin 1: assessment of interleukin 6 and tumor necrosis factor alpha induction in vivo. The Cancer Research Campaign Phase I Committee. J. Natl. Cancer Inst. 1993; 85, 1812–1818.

218. Mohr H., Pettit G. R., Plessing-Menze A. Co-induction of lymphokine synthesis by the antineoplastic bryostatins. Immunobiology 1987; 175, 420–430.

219. Alkon D. L., Sun M-K., Nelson T. J. PKC signaling deficits: a mechanistic hypothesis for the origins of Alzheimer’s disease. Trends Pharmacol. Sci. 2007; 28, 51–60.

220. Sun M-K., Alkon D. L. The ‘memory kinases’: roles of PKC isoforms in signal processing and memory formation. Prog. Mol. Biol. Transl. Sci. 2014; 122, 31–59.

221. Pandey G. N., Dwivedi Y. Focus on protein kinase A and protein kinase C, critical components of signal transduction system, in mood disorders and suicide. Int. J. Neuropsychopharmacol. 2005; 8, 1–4.

222. Shelton R. C. The molecular neurobiology of depression. Psychiatr. Clin. North Am. 2007; 30, 1–11.

223. Sun M-K., Alkon D. L. Dual effects of bryostatin-1 on spatial memory and depression. Eur. J. Pharmacol. 2005; 512, 43–51.

224. NCT02431468. A Study Assessing Bryostatin in the Treatment of Moderately Severe to Severe Alzheimer’s Disease. https://clinicaltrials.gov/ct2/show/NCT02431468 (30. 5. 2017).

225. Mehla R., Bivalkar-Mehla S., Zhang R., Handy I., Albrecht H., Giri S., Nagarkatti P., Nagarkatti M., Chauhan A. Bryostatin modulates latent HIV-1 infection via PKC and AMPK signaling but inhibits acute infection in a receptor independent manner. PloS One 2010; 5, e11160.

226. Díaz L., Martínez-Bonet M., Sánchez J., Fernández-Pineda A., Jiménez J. L., Muñoz E., Moreno S., Álvarez S., Muñoz-Fernández M. Á. Bryostatin activates HIV-1 latent expression in human astrocytes through a PKC and NF-ĸB-dependent mechanism. Sci. Rep. 2015; 5, 12442.

227. Boto W. M., Brown L., Chrest J., Adler W. H. Distinct modulatory effects of bryostatin 1 and staurosporine on the biosynthesis and expression of the HIV receptor protein (CD4) by T cells. Cell Regul. 1991; 2, 95–103.

228. Gutiérrez C., Serrano-Villar S., Madrid-Elena N., Pérez-Elías M. J., Martín M. E., Barbas C., Ruipérez J., Muñoz E., Muñoz-Fernández M. A., Castor T., Moreno S. Bryostatin-1 for latent virus reactivation in HIV-infected patients on antiretroviral therapy. AIDS Lond. Engl. 2016; 30, 1385–1392.

229. Ajani J. A., Jiang Y., Faust J., Chang B. B., Ho L., Yao J. C., Rousey S., Dakhil S., Cherny R. C., Craig C., Bleyer A. 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 2006; 24, 353–357.

230. Barr P. M., Lazarus H. M., Cooper B. W., Schluchter M. D., Panneerselvam A., Jacobberger J. W., Hsu J. W., Janakiraman N., Simic A., Dowlati A., Remick S. C. Phase II study of bryostatin 1 and vincristine for aggressive non-Hodgkin lymphoma relapsing after an autologous stem cell transplant. Am. J. Hematol. 2009; 84, 484–487.

231. Lam A. P., Sparano J. A., Vinciguerra V., Ocean A. J., Christos P., Hochster H., Camacho F., Goel S., Mani S., Kaubisch A. Phase II study of paclitaxel plus the protein kinase C inhibitor bryostatin-1 in advanced pancreatic carcinoma. Am. J. Clin. Oncol. 2010; 33, 121–124.

232. Nezhat F., Wadler S., Muggia F., Mandeli J., Goldberg G., Rahaman J., Runowicz C., Murgo A. J., Gardner G. J. Phase II trial of the combination of bryostatin-1 and cisplatin in advanced or recurrent carcinoma of the cervix: a New York Gynecologic Oncology Group study. Gynecol. Oncol. 2004; 93, 144–148.

233. DeChristopher B. A., Fan A. C., Felsher D. W., Wender P. A. ‘Picolog’, a synthetically-available bryostatin analog, inhibits growth of MYC-induced lymphoma in vivo. Oncotarget 2012; 3, 58–66.

234. Tan Z., Turner R. C., Leon R. L., Li X., Hongpaisan J., Zheng W., Logsdon A. F., Naser Z. J., Alkon D. L., Rosen C. L., Huber J. D. Bryostatin improves survival and reduces ischemic brain injury in aged rats after acute ischemic stroke. Stroke 2013; 44, 3490–3497.

235. Tan Z., Lucke-Wold B. P., Logsdon A. F., Turner R. C., Tan C., Li X., Hongpaison J., Alkon D. L., Simpkins J. W., Rosen C. L., Huber J. D. Bryostatin extends tPA time window to 6 h following middle cerebral artery occlusion in aged female rats. Eur. J. Pharmacol. 2015; 764, 404–412.

236. Sun M.-K., Hongpaisan J., Alkon D. L. Postischemic PKC activation rescues retrograde and anterograde long-term memory. Proc. Natl. Acad. Sci. U. S. A. 2009; 106, 14676–14680.

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

238. Fairchild C. R., Johnston K. J., Peterson R. W., Cornell L. A., Bifario M., Raventos-Suarez C., et al. Halimide, a novel cytotoxic marine natural product, destabilizes microtubules and demonstrates in vivo antitumor activity. Proc Am Cancer Res 1998; 165.

239. Kanoh K., Kohno S., Asari T., Harada T., Katada J. Phenylahistin: a new mammalian cell cycle inhibitor produced by Aspergillus ustus. Bioorg Med Chem Lett 1997; 2847–2852.

240. Mita M. M., Spear M. A., Yee L. K., Mita A. C., Heath E. I., Papadopoulos K. P., Federico K. C., Reich S. D., Romero O., Malburg L., Pilat M., Lloyd G. K., Neuteboom S. T. C., Cropp G., Ashton E., LoRusso P. M. Phase 1 first-in-human trial of the vascular disrupting agent plinabulin(NPI-2358) in patients with solid tumors or lymphomas. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2010; 16, 5892–5899.

241. Millward M., Mainwaring P., Mita A., Federico K., Lloyd G. K., Reddinger N., Nawrocki S., Mita M., Spear M. A. Phase 1 study of the novel vascular disrupting agent plinabulin (NPI-2358) and docetaxel. Invest. New Drugs 2012; 30, 1065–1073.

242. Feling R. H., Buchanan G. O., Mincer T. J., Kauffman C. A., Jensen P. R., Fenical W. Salinosporamide A: a highly cytotoxic proteasome inhibitor from a novel microbial source, a marine bacterium of the new genus salinospora. Angew. Chem. Int. Ed Engl. 2003; 42, 355–357.

243. King R. W., Deshaies R. J., Peters J. M., Kirschner M. W. How proteolysis drives the cell cycle. Science 1996; 274, 1652–1659.

244. Levin N., Spencer A., Harrison S. J., Chauhan D., Burrows F. J., Anderson K. C., Reich S. D., Richardson P. G., Trikha M. Marizomib irreversibly inhibits proteasome to overcome compensatory hyperactivation in multiple myeloma and solid tumour patients. Br. J. Haematol. 2016; 174, 711–720.

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

246. Ahn K. S., Sethi G., Chao T.-H., Neuteboom S. T. C., Chaturvedi M. M., Palladino M. A., Younes A., Aggarwal B. B. Salinosporamide A (NPI-0052) potentiates apoptosis, suppresses osteoclastogenesis, and inhibits invasion through down-modulation of NF-kappaB regulated gene products. Blood 2007; 110, 2286–2295.

247. Bassères D. S., Baldwin A. S. Nuclear factor-kappaB and inhibitor of kappaB kinase pathways in oncogenic initiation and progression. Oncogene 2006; 25, 6817–6830.

248. Hideshima T., Ikeda H., Chauhan D., Okawa Y., Raje N., Podar K., Mitsiades C., Munshi N. C., Richardson P. G., Carrasco R. D., Anderson K. C. Bortezomib induces canonical nuclear factor-kappaB activation in multiple myeloma cells. Blood 2009; 114, 1046–1052.

249. Cusack J. C., Liu R., Xia L., Chao T-H., Pien C., Niu W., Palombella V. J., Neuteboom S. T. C., Palladino M. A. NPI-0052 enhances tumoricidal response to conventional cancer therapy in a colon cancer model. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006; 12, 6758–6764.

250. Ruiz S., Krupnik Y., Keating M., Chandra J., Palladino M., McConkey D. The proteasome inhibitor NPI-0052 is a more effective inducer of apoptosis than bortezomib in lymphocytes from patients with chronic lymphocytic leukemia. Mol. Cancer Ther. 2006; 5, 1836–1843.

251. Miller C. P., Ban K., Dujka M. E., McConkey D. J., Munsell M., Palladino M., Chandra J. NPI-0052, a novel proteasome inhibitor, induces caspase-8 and ROS-dependent apoptosis alone and in combination with HDAC inhibitors in leukemia cells. Blood 2007; 110, 267–277.

252. Millward M., Price T., Townsend A., Sweeney C., Spencer A., Sukumaran S., Longenecker A., Lee L., Lay A., Sharma G., Gemmill R. M., Drabkin H. A., Lloyd G. K., Neuteboom S. T. C., McConkey D. J., Palladino M. A., Spear M. A. Phase 1 clinical trial of the novel proteasome inhibitor marizomib with the histone deacetylase inhibitor vorinostat in patients with melanoma, pancreatic and lung cancer based on in vitro assessments of the combination. Invest. New Drugs 2012; 30, 2303–2317.

253. Richardson P. G., Zimmerman T. M., Hofmeister C. C., Talpaz M., Chanan-Khan A. A., Kaufman J. L., Laubach J. P., Chauhan D., Jakubowiak A. J., Reich S., Trikha M., Anderson K. C. Phase 1 study of marizomib in relapsed or relapsed and refractory multiple myeloma: NPI-0052-101 Part 1. Blood 2016; 127, 2693–2700.

254. Harrison S. J., Mainwaring P., Price T., Millward M. J., Padrik P., Underhill C. R., Cannell P. K., Reich S. D., Trikha M., Spencer A. Phase I Clinical Trial of Marizomib (NPI-0052) in Patients with Advanced Malignancies Including Multiple Myeloma: Study NPI-0052-102 Final Results. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016; 22, 4559–4566.

255. NCT00461045. Phase 1/2 Clinical Trial of NPI-0052 in Patients With Relapsed or Relapsed/Refractory Multiple Myeloma. https://clinicaltrials.gov/ct2/show/NCT00461045 (30. 5. 2017).

256. NCT02330562. Stage 1: Marizomib + Bevacizumab in WHO Gr IV GBM; Stage 2: Marizomib Alone. https://clinicaltrials.gov/ct2/show/NCT02330562 (30. 5. 2017).

257. NCT02903069. Study of Marizomib With Temozolomide and Radiotherapy in Patients With Newly Diagnosed Brain Cancer. https://clinicaltrials.gov/ct2/show/NCT02903069 (30. 5. 2017).

Labels
Pharmacy Clinical pharmacology
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#