Anti-tumour drugs of marine origin currently at various stages of clinical trials (review)

  • E. A. Bocharova A. O. Kovalevsky Institute of Biology of the Southern Seas of RAS
  • N. I. Kopytina A. O. Kovalevsky Institute of Biology of the Southern Seas of RAS
  • Е. Е. Slynko Yaroslavl State Agricultural Academy
Keywords: cancer; marine organisms; preparations; cytotoxic preparations; clinical studies

Abstract

Oncological diseases for a long time have remained one of the most significant health problems of modern society, which causes great losses in its labour and vital potential. Contemporary oncology still faces unsolved issues as insufficient efficacy of treatment of progressing and metastatic cancer, chemoresistance, and side-effects of the traditional therapy which lead to disabilities among or death of a high number of patients. Development of new anti-tumour preparations with a broad range of pharmaceutical properties and low toxicity is becoming increasingly relevant every year. The objective of the study was to provide a review of the recent data about anti-tumour preparations of marine origin currently being at various phases of clinical trials in order to present the biological value of marine organisms – producers of cytotoxic compounds, and the perspectives of their use in modern biomedical technologies. Unlike the synthetic oncological preparations, natural compounds are safer, have broader range of cytotoxic activity, can inhibit the processes of tumour development and metastasis, and at the same time have effects on several etiopathogenic links of carcinogenesis. Currently, practical oncology uses 12 anti-tumour preparations of marine origin (Fludarabine, Cytarabine, Midostaurin, Nelarabine, Eribulin mesylate, Brentuximab vedotin, Trabectedin, Plitidepsin, Enfortumab vedotin, Polatuzumab vedotin, Belantamab mafodotin, Lurbinectedin), 27 substances are at different stages of clinical trials. Contemporary approaches to the treatment of oncological diseases are based on targeted methods such as immune and genetic therapies, antibody-drug conjugates, nanoparticles of biopolymers, and metals. All those methods employ bioactive compounds of marine origin. Numerous literature data from recent years indicate heightened attention to the marine pharmacology and the high potential of marine organisms for the biomedicinal and pharmaceutic industries.

References

Abidizadegan, M., Peltomaa, E., & Blomster, J. (2021). The potential of cryptophyte algae in biomedical and pharmaceutical applications. Frontiers in Pharmacology, 11, 618836.

Advani, R. H., Lebovic, D., Chen, A., Brunvand, M., Goy, A., Chang, J., Hochberg, E., Yalamanchili, S., Kahn, R., Lu, D., Agarwal, P., Dere, R. C., Hsieh, H.-J., Jones, S., Chu, Y.-W., & Cheson, B. D. (2017). Phase I study of the anti-CD22 antibody-drug conjugate pinatuzumab vedotin with/without rituximab in patients with relapsed/refractory B-cell non-Hodgkin’s lymphoma. Clinical Cancer Research, 23(5), 1167–1176.

Agrawal, S., Adholeya, A., & Deshmukh, S. K. (2016) The pharmacological potential of non-ribosomal peptides from marine sponge and tunicates. Frontiers in Pharmacology, 7, 333.

Ahmad, T. B., Liu, L., Kotiw, M., & Benkendorff, K. (2018). Review of anti-inflammatory, immune-modulatory and wound healing properties of molluscs. Journal of Ethnopharmacology, 210, 156–178.

Aiub, C., Giannerini, A., Ferreira, F., Mazzei, J., Stankevicins, L., Lobo-Hajdu, G., Guimaraes, P., Hajdu, E., & Felzenszwalb, I. (2006). Genotoxic evaluation of extracts from Aplysina fulva, a brazilian marine sponge. Mutation Research, 611, 34–41.

Akashi, Y., Okamoto, I., Suzuki, M., Tamura, K., Iwasa, T., Hisada, S., Satoh, T., Nakagawa, K., Ono, K., & Fukuoka, M. (2007). The novel microtubule-interfering agent TZT-1027 enhances the anticancer effect of radiation in vitro and in vivo. British Journal of Cancer, 96(10), 1532–1539.

Akla, B., Broussas, M., Loukili, N., Robert, A., Beau-Larvor, C., Malissard, M., Boute, N., Champion, T., Haeuw, J.-F., Beck, A., Perez, M., Dreyfus, C., Pavlyuk, M., Chetaille, E., & Corvaia, N. (2020). Efficacy of the antibody-drug conjugate W0101 in preclinical models of IGF-1 receptor overexpressing solid tumors. Molecular Cancer Therapeutics, 19(1), 168–177.

Almhanna, K., Wright, D., Mercade, T. M., Van Laethem, J.-L., Gracian, A. C., Guillen-Ponce, C., Faris, J., Lopez, C. M., Hubner, R. A., Bendell, J., Bols, A., Feliu, J., Starling, N., Enzinger, P., Mahalingham, D., Messersmith, W., Yang, H., Fasanmade, A., Danaee, H., & Kalebic, T. (2017). A phase II study of antibody-drug conjugate, TAK-264 (MLN0264) in previously treated patients with advanced or metastatic pancreatic adenocarcinoma expressing guanylyl cyclase. Investigational New Drugs, 35(5), 634–641.

Al-Musawi, S., Albukhaty, S., Al-Karagoly, H., & Almalki, F. (2020). Design and synthesis of multi-functional fsuperparamagnetic core-gold shell coated with chitosan and folate nanoparticles for targeted antitumor therapy. Nanomaterials, 11(1), 32.

Alonso-Álvarez, S., Pardal, E., Sánchez-Nieto, D., Navarro, M., Caballero, M. D., Mateos, M. V., & Martín, A. (2017). Plitidepsin: Design, development, and potential place in therapy. Drug Design Development and Therapy, 11, 253–264.

Alves, A. J. S., Pereira, J. A., Dethoup, T., Cravo, S., Mistry, S., Silva, A. M. S., Pinto, M. M. M., & Kijjoa, A. A. (2019). New meroterpene, a new benzofuran derivative and other constituents from cultures of the marine sponge-associated fungus Acremonium persicinum KUFA 1007 and their anticholinesterase activities. Marine Drugs, 17(6), 379.

Amaya, M. L., Jimeno, A., & Kamdar, M. (2020). Polatuzumab vedotin to treat relapsed or refractory diffuse large B-cell lymphoma, in combination with bendamustine plus rituximab. Drugs Today, 56(4), 287–294.

Andel, L. V., Rosing, H., Schellens, J. H. M., & Beijnen, J. H. (2018). Review of chromatographic bioanalytical assays for the quantitative determination of marine-derived drugs for cancer treatment. Marine Drugs, 16(7), 246.

Anderson, B. L., Teyton, L., Bendelac, A., & Savage, P. B. (2013). Stimulation of natural killer T cells by glycolipids. Molecules, 18(12), 15662–15688.

Arumugam, V., Venkatesan, M., Ramachandran, S., & Sundaresan, U. (2018). Bioactive peptides from marine ascidians and future drug development – a review. International Journal of Peptide Research and Therapeutics, 24(1), 13–18.

Autio, K. A., Boni, V., Humphrey, R. W., & Naing, A. (2020). Probody therapeutics: An emerging class of therapies designed to enhance on-target effects with reduced off-tumor toxicity for use in immuno-oncology. Clinical Cancer Research, 26(5), 984–989.

Avila, C., & Angulo-Preckler, C. (2020). Bioactive compounds from marine heterobranchs. Marine Drugs, 18(12), 657.

Babu, A., & Ramesh, R. (2017). Multifaceted applications of chitosan in cancer drug delivery and therapy. Marine Drugs, 15(4), 96.

Balasubramaniam, V., Gunasegavan, R. D.-N., Mustar, S. S., Lee, J. C., & Noh, M. F. M. (2021). Isolation of industrial important bioactive compounds from microalgae. Molecules, 26(4), 943.

Barok, M., Puhka, M., Yazdi, N., & Joensuu, H. (2021). Extracellular vesicles as modifiers of antibody drug conjugate efficacy. Journal Extracell Vesicles, 10(4), e12070.

Barreca, M., Spanò, V., Montalbano, A., Cueto, M., Marrero, A. R. D., Deniz, I., Erdoğan, A., Bilela, L. L., Moulin, C., Taffin-de-Givenchy, E., Spriano, F., Perale, G., Mehiri, M., Rotter, A., Thomas, O. P., Barraja, P., Gaudêncio, S. P., & Bertoni, F. (2020). Marine anticancer agents: An overview with a particular focus on their chemical classes. Marine Drugs, 18(12), 619.

Belgiovine, C., Bello, E., Liguori, M., Craparotta, I., Mannarino, L., Paracchini, L., Beltrame, L., Marchini, S., Galmarini, C. M., Mantovani, A., Frapolli, R., Allavena, P., & D’Incalci, M. (2017). Lurbinectedin reduces tumour-associated macrophages and the inflammatory tumour microenvironment in preclinical models. British Journal of Cancer, 117(5), 628–638.

Bibi, F., Faheem, M., Azhar, E. I., Yasir, M., Alvi, S. A., Kamal, M. A., Ullah, I., & Naseer, M. I. (2017). Bacteria from marine sponges: A source of new drugs. Current Drug Metabolism, 18(1), 11–15.

Biteghe, N. F. A., Mungra, N., Toung, C. N. E., De La Croix, N. J., Engohang-Ndong, J., Vignaux, G., Padayachee, E., Naran, K., & Barth, S. (2020). Advances in epidermal growth factor receptor specific immunotherapy: Lessons to be learned from armed antibodies. Oncotarget, 11(38), 3531–3557.

Blackall, L. L., Wilson, B., & Van Oppen, M. J. H. (2015). Coral – the world’s most diverse symbiotic ecosystem. Molecular Ecology, 24(21), 5330–5347.

Blay, J. Y., Blay, J. Y., Pápai, Z., Tolcher, A. W., Italiano, A., Cupissol, D., López-Pousa, A., Chawla, S. P., Bompas, E., Babovic, N., Penel, N., Isambert, N., Staddon, A. P., Saâda-Bouzid, E., Santoro, A., Franke, F. A., Cohen, P., Le-Guennec, S., & Demetri, G. (2015). Ombrabulin plus cisplatin versus placebo plus cisplatin in patients with advanced soft-tissue sarcomas after failure of anthracycline and ifosfamide chemotherapy: A randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet Oncology, 16(5), 531–540.

Boekhoven, J., Zha, R. H., Tantakitti, F., Zhuang, E., Zandi, R., Newcomb, C. J., & Stupp, S. I. (2015). Alginate-peptide amphiphile core-shell microparticles as a targeted drug delivery system. RSC Advances, 5(12), 8753–8756.

Boni, V., Meisel, J. L., Tkaczuk, K. H., Wang, Z., Alemany, C., Sterrenberg, D., Wang, Y., Wang, Z., & Han, H. S. (2019). 196TiPSGNLVA-002: Single arm, open label, phase Ib/II study of ladiratuzumab vedotin (LV) in combination with pembrolizumab for first-line treatment of patients with unresectable locally-advanced or metastatic triple-negative breast cancer. Annals of Oncology, 30(3), 63.

Boni, V., Sharma, M. R., & Patnaik, A. (2020). The resurgence of antibody drug conjugates in cancer therapeutics: Novel targets and payloads. American Society of Clinical Oncology Educational Book, 40, 1–17.

Bourillon, L., Bourgier, C., Gaborit, N., Garambois, V., Llès, E., Zampieri, A., Ogier, C., Jarlier, M., Radosevic-Robin, N., Orsetti, B., Delpech, H., Theillet, C., Colombo, P. E., Azria, D., Pèlegrin, A., Larbouret, C., & Chardès, T. (2019). An auristatin-based antibody-drug conjugate targeting HER3 enhances the radiation response in pancreatic cancer. International Journal Cancer, 145(7), 1838–1851.

Calcabrini, C., Catanzaro, E., Bishayee, A., Turrini, E., & Fimognari, C. (2017). Marine sponge natural products with anticancer potential: An updated review. Marine Drugs, 15(10), 310.

Calvo, E., Moreno, V., Flynn, M., Holgado, E., Olmedo, M. E., Lopez Criado, M. P., Kahatt, C., Lopez-Vilariño, J. A., Siguero, M., Fernandez-Teruel, C., Cullell-Young, M., Matos-Pita, A. S., & Forster, M. (2017). Antitumor activity of lurbinectedin (PM01183) and doxorubicin in relapsed small-cell lung cancer: Results from a phase I study. Annals of Oncology, 28(10), 2559–2566.

Carroll, A. R., Copp, B. R., Davis, R. A., Keyzers, R. A., & Prinsep, M. R. (2020). Marine natural products. Natural Product Reports, 37, 175–223.

Chakraborty, K., & Joy, M. (2020). High-value compounds from the molluscs of marine and estuarine ecosystems as prospective functional food ingredients: An overview. Food Research International, 137, 109637.

Chen, R., Herrera, A. F., Hou, J., Chen, L., Wu, J., Guo, Y., Synold, T. W., Ngo, V. N., Puverel, S., Mei, M., Popplewell, L., Yi, S., Song, J. Y., Tao, S., Wu, X., Chan, W. C., Forman, S. J., Kwak, L. W., Rosen, S. T., & Newman, E. M. (2020). Inhibition of MDR1 overcomes resistance to brentuximab vedotin in Hodgkin lymphoma. Clinical Cancer Research, 26(5), 1034–1044.

Cheng, M.-M., Tang, X.-T., Sun, Y.-T., Song, D.-Y., Cheng, Y.-J., Liu, H., Li, P.-L., & Li, G.-Q. (2020). Biological and chemical diversity of marine sponge-derived microorganisms over the last two decades from 1998 to 2017. Molecules, 25(4), 853.

Cheng, X., Li, J., Tanaka, K., Majumder, U., Milinichik, A. Z., Verdi, A. C., Maddage, C. J., Rybinski, K. A., Fernando, S., Fernando, D., Kuc, M., Furuuchi, K., Fang, F., Uenaka, T., Grasso, L., & Albone, E. F. (2018). MORAb-202, an antibody-drug conjugate utilizing humanized anti-human FRα farletuzumab and the microtubule-targeting agent eribulin, has potent antitumor activity. Molecular Cancer Therapeutics, 17(12), 2665–2675.

Cheung, R. C. F., Ng, T. B., & Wong, J. H. (2015). Marine peptides: Bioactivities and applications. Marine Drugs, 13(7), 4006–4043.

Choi, M. Y., Widhopf, G. F., Ghia, E. M., Kidwell, R. L., Hasan, M. K., Yu, J., Rassenti, L. Z., Chen, L., Chen, Y., Pittman, E., Pu, M., Messer, K., Prussak, C. P., Castro, J. E., Jamieson, C., & Kipps, T. J. (2018). Phase I trial: Cirmtuzumab inhibits ROR1 signaling and stemness signatures in patients with chronic lymphocytic leukemia. Cell Stem Cell, 22(6), 951–959.

Choi, M. Y., Widhopf, G. F., Wu, C. C. N., Cui, B., Lao, F., Sadarangani, A., Cavagnaro, J., Prussak, C., Carson, D. A., Jamieson, C., & Kipps, T. J. (2015). Pre-clinical specificity and safety of UC-961, a first-in-class monoclonal antibody targeting ROR1. Clinical Lymphoma, Myeloma and Leukemia, 15, 167–169.

Choi, Y., & Diefenbach, C. S. (2020). Polatuzumab vedotin: A new target for B cell malignancies. Current Hematologic Malignancy Reports, 15, 125–129.

Ciavatta, M. L., Lefranc, F., Carbon, M., Mollo, E., Gavagnin, M., Betancourt, T., Dasari, R., Kornienko, A., & Kiss, R. (2017). Marine mollusk-derived agents with antiproliferative activity as promising anticancer agents to overcome chemotherapy resistance. Medicinal Research Reviews, 37(4), 702–801.

Ciavatta, M. L., Lefranc, F., Vieira, L. M., Kiss, R., Carbone, M., Van Otterlo, W. A. L., Lopanik, N. B., & Waeschenbach, A. (2020). The phylum Bryozoa: From biology to biomedical potential. Marine Drugs, 18(4), 200.

Cooper, T. M., Absalon, M. J., Alonzo, T. A., Gerbing, R. B., Leger, K. J., Hirsch, B. A., Pollard, J., Razzouk, B. I., Aplenc, R., & Kolb, E. A. (2020). Phase I/II study of CPX-351 followed by fludarabine, cytarabine, and granulocyte-colony stimulating factor for children with relapsed acute myeloid leukemia: A report from the children’s oncology group. Journal of Clinical Oncology, 38(19), 2170–2177.

Cragg, G. M., Kingston, D. G. I., & Newman, D. J. (2012). The dolastatins: Novel antitumor agents from Dolabella auricularia. Anticancer Agents from Natural Products. Boca Raton, CRC Press.

De Bono, J. S., Concin, N., Hong, D. S., Thistlethwaite, F. C., Machiels, J.-P., Arkenau, H. T., Plummer, R., Jones, R. H., Nielsen, D., Windfeld, K., Ghatta, S., Slomovitz, B. M., Spicer, J. F., Yachnin, J., Ang, J. E., Mau-Sørensen, P. M., Forster, M. D., Collins, D., Dean, E., Rangwala, R. A., & Lassen, U. L. (2019). Tisotumab vedotin in patients with advanced or metastatic solid tumours (InnovaTV 201): A first-in-human, multicentre, phase 1-2 trial. Lancet Oncology, 20(3), 383–393.

De Martel, C., Georges, D., Bray, F., Ferlay, J., & Clifford, G. M. (2020). Global burden of cancer attributable to infections in 2018: A worldwide incidence analysis. Lancet Global Health, 8(2), 180–190.

Demay, J., Halary, S., Knittel-Obrecht, A., Villa, P., Duval, C., Hamlaoui, S., Roussel, T., Yéprémian, C., Reinhardt, A., Bernard, C., & Marie, B. (2021). Anti-inflammatory, antioxidant, and wound-healing properties of cyanobacteria from thermal mud of Balaruc-Les-Bains, France: A multi-approach study. Biomolecules, 11(1), 28.

Deshmukh, S. K., Prakash, V., & Ranjan, N. (2018). Marine fungi: A source of potential anticancer compounds. Frontiers in Microbiology, 8, 2536.

Diaa, T. A. Y., Almagthali, H., Shaala, L. A., & Schmidt, E. W. (2020). Secondary metabolites of the genus Didemnum: A comprehensive review of chemical diversity and pharmacological properties. Marine Drugs, 18(6), 307.

Dou, D., & Dong, B. (2019). Origins and bioactivities of natural compounds derived from marine ascidians and their symbionts. Marine Drugs, 17(12), 670.

Dunsmore, K. P., Winter, S. S., Devidas, M., Wood, B. L., Esiashvili, N., Chen, Z., Eisenberg, N., Briegel, N., Hayashi, R. J., Gastier-Foster, J. M., Carroll, A. J., Heerema, N. A., Asselin, B. L., Rabin, K. R., Zweidler-Mckay, P. A., Raetz, E. A., Loh, M. L., Schultz, K. R., Winick, N. J., Carroll, W. L., & Hunger, S. P. (2020). Children’s oncology group AALL0434: A phase III randomized clinical trial testing nelarabine in newly diagnosed T-cell acute lymphoblastic leukemia. Journal of Clinical Oncology, 38(28), 3282–3293.

Dyshlovoy, S. A., & Honecker, F. (2015). Marine compounds and cancer: Where do we stand? Marine Drugs, 13(9), 5657–5665.

Dyshlovoy, S. A., & Honecker, F. (2020). Marine compounds and cancer: Updates. Marine Drugs, 18(12), 643.

Eastman, A. (2017). Improving anticancer drug development begins with cell culture: Misinformation perpetrated by the misuse of cytotoxicity assays. Oncotarget, 8(5), 8854–8866.

El Bairi, K., Amrani, M., Kandhro, A. H., Kandhro, A. H. & Afqir, S. (2017). Prediction of therapy response in ovarian cancer: Where are we now? Critical Reviews in Clinical Laboratory Sciences, 54(4), 233–266.

El-Hossary, E. M., Abdel-Halim, M., Ibrahim, E. S., Pimentel-Elardo, S. M., Nodwell, J. R., Handoussa, H., Abdelwahab, M., Holzgrabe, U., & Abdelmohsen, U. R. (2020). Natural products repertoire of the Red Sea. Marine Drugs, 18(9), 457.

El-Kashef, D. H., Youssef, F. S., Reimche, I., Teusch, N., Müller, W. E. G., Lin, W., Frank, M., Liu, Z., & Proksch, P. (2021). Polyketides from the marine-derived fungus Aspergillus falconensis: In silico and in vitro cytotoxicity studies. Bioorganic and Medicinal Chemistry, 29, 115883.

Elliot, A., Myllymäki, H., & Feng, Y. (2020). Inflammatory responses during tumour initiation: From zebrafish transgenic models of cancer to evidence from mouse and man. Cells, 9(4), 1018.

Ercolano, G., De Cicco, P., & Ianaro, A. (2019). New drugs from the sea: Pro-apoptotic activity of sponges and algae derived compounds. Marine Drugs, 17(1), 31.

Esmaeelian, B., Benkendorff, K., Leu, R. K. L., & Abbott, C. A. (2018). Simultaneous assessment of the efficacy and toxicity of marine mollusc-derived brominated indoles in an in vivo model for early stage colon cancer. Integrative Cancer Therapies, 17(2), 248–262.

Evidente, A., Kornienko, A. Cimmino, A., Andolfi, A., Lefranc, F., Mathieu, V., & Kiss, R. (2014). Fungal metabolites with anticancer activity. Natural Product Reports, 31, 617–627.

Fadia, S. Y., & Singab, A. N. B. (2021). An updated review on the secondary metabolites and biological activities of Aspergillus ruber and Aspergillus flavus and exploring the cytotoxic potential of their isolated compounds using virtual screening. Evid Based Complementary and Alternative Medicine, 2021, 8860784.

Fang, W.-Y., Dahiya, R., Qin, H.-L., Mourya, R., & Maharaj, S. (2016). Natural proline-rich cyclopolypeptides from marine organisms: Chemistry, synthetic methodologies and biological status. Marine Drugs, 14(11), 194.

Ferlay, J., Colombet, M., Soerjomataram, I., Mathers, C., Parkin, D. M., Piñeros, M., Znaor, A., & Bray, F. (2019). Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. International Journal of Cancer, 144(8), 1941–1953.

Fidler, M. M., Bray, F., & Soerjomataram, I. (2018). The global cancer burden and human development: A review. Scandinavian Journal of Public Health, 46(1), 27–36.

Fitton, J. H., Park, A. Y., Karpiniec, S. S., & Stringer, D. N. (2020). Fucoidan and lung function: Value in viral infection. Marine Drugs, 19(1), 4.

Fitton, J. H., Stringer, D. N., Park, A. Y., & Karpiniec, S. S. (2019). Therapies from fucoidan: New developments. Marine Drugs, 17(10), 571.

Forero-Torres, A., Kolibaba, K. S., Lamy, T., Jones, S., Biostat, M. S., Lee, C., & Sharman, J. (2016). Polatuzumab vedotin combined with obinutuzumab, cyclophosphamide, doxorubicin, and prednisone (G-CHP) for patients with previously untreated diffuse large B-cell lymphoma (DLBCL): Preliminary results of a phase Ib/II dose-escalation study. Blood, 128(22), 1856.

Francisco, J. A., Cerveny, C. G., & Meyer, D. L. (2003). cAC10-vcMMAE, an anti-CD30-monomethyl auristatin E conjugate with potent and selective antitumor activity. Blood, 102(4), 1458–1465.

Frankel, B. M., Cachia, D., Patel, S. J., & Das, A. (2020). Targeting subventricular zone progenitor cells with intraventricular liposomal encapsulated cytarabine in patients with secondary glioblastoma: A report of two cases. SN Comprehensive Clinical Medicine, 2(6), 836–843.

Frau J., Flores-Holguín, N., & Glossman-Mitnik, D. (2019). Chemical reactivity theory and empirical bioactivity scores as computational peptidology alternative tools for the Study of two anticancer peptides of marine origin. Molecules, 24(6), 1115.

Fuji, N., Ueda, Y., Fujiwara, H., Toh, T., Yoshimura, T., & Yamagishi, H. (2000). Antitumor effect of alpha-galactosylceramide (KRN7000) on spontaneous hepatic metastases requires endogenous interleukin 12 in the liver. Clinical Cancer Research, 6(8), 3380–3387.

Gagliardi, A., Giuliano, E., Venkateswararao, E., Fresta, M., Bulotta, S., Awasthi, V., & Cosco, D. (2021). Biodegradable polymeric nanoparticles for drug delivery to solid tumors. Frontiers in Pharmacology, 12, 601626.

Galasso, C., Gentile, A., Orefice, I., Ianora, A., Bruno, A., Noonan, D. M., Sansone, C., Albini, A., & Brunet, C. (2019). Microalgal derivatives as potential nutraceutical and food supplements for human health: A focus on cancer prevention and interception. Nutrients, 11(6), 1226.

Gao, J., & Hamann, M. T. (2011). Chemistry and biology of kahalalides. Chemical Reviews, 111(5), 3208–3235.

Gavriilidou, A., Mackenzie, T. A. M., Sánchez, P., Tormo, J. R., Ingham, C., Smidt, H., & Sipkema, D. (2021). Bioactivity screening and gene-trait matching across marine sponge-associated bacteria. Marine Drugs, 19(2), 75.

Gentile, D., Patamia, V., Scala, A., Sciortino, M. T., Piperno, A., & Rescifina, A. (2020). Putative inhibitors of SARS-CoV-2 main protease from a library of marine natural products: A virtual screening and molecular modeling study. Marine Drugs, 2021(19), 64.

George, A., Shah, P. A., & Shrivastav, P. S. (2019). Natural biodegradable polymers based nano-formulations for drug delivery: A review. International Journal of Pharmaceutics, 561, 244–264.

Gerber, D. E., Infante, J. R., Gordon, M. S., Goldberg, S. B., Martín, M., Felip, E., Martinez, G. M, Schiller, J. H., Spigel, D. R., Cordova, J., Westcott, V., Wang, Y., Shames, D. S., Choi, Y., Kahn, R., Dere, R. C., Samineni, D., Xu, J., Lin, K., Wood, K., Royer-Joo, S., Lemahieu, V., Schuth, E., Vaze, A., Maslyar, D., Humke, E. W., & Burris, H. A. (2020). 3rd phase Ia study of anti-NaPi2b antibody-drug conjugate lifastuzumab vedotin DNIB0600A in patients with non-small cell lung cancer and platinum-resistant ovarian cancer. Clinical Cancer Research, 26(2), 364–372.

Gerwick, W. H., & Moore, B. S. (2012). Lessons from the past and charting the future of marine natural products drug discovery and chemical biology. Journal of Chemical Biology, 19(1), 85–98.

Gnanambal, K. M., & Lakshmipathy, S. V. (2016). Dictyoceratidan poisons: Defined mark on microtubule-tubulin dynamics. Life Sciences, 148, 229–240.

Gogineni, V. M., & Hamann, M. T. (2018). Marine natural product peptides with therapeutic potential: Chemistry, biosynthesis, and pharmacology. Biochimica et Biophysica Acta, 1862(1), 81–196.

Golan, T., Khvalevsky, E. Z., Hubert, A., Gabai, R. M., Hen, N., Segal, A., Domb, A., Harari, G., David, E. B., Raskin, S., Goldes, Y., Goldin, E., Eliakim, R., Lahav, M., Kopleman, Y., Dancour, A., Shemi, A., & Galun, E. (2015). RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget, 6(27), 24560–24570.

Gomes, N. G. M., Dasari, R., Chandra, S., Kiss, R., & Kornienko, A. (2016). Marine invertebrate metabolites with anticancer activities: Solutions to the “Supply problem”. Marine Drugs, 14(5), 98.

Gray, B. P., Song, X., Hsu, D. S., Kratschmer, C., Levy, M., Barry, A. P., & Sullenger, B. A. (2020). An aptamer for broad cancer targeting and therapy. Cancers, 12(11), 3217.

Graziani, E. I., Sung, M., Ma, D., Narayanan, B., Marquette, K., Puthenveetil, S., Tumey, L. N., Bikker, J., Casavant, J., Bennett, E. M., Charati, M. B., Golas, J., Hosselet, C., Rohde, C. M., Hu, G., Guffroy, M., Falahatpisheh, H., Finkelstein, M., Clark, T., Barletta, F., Tchistiakova, L., Lucas, J., Rosfjord, E., Loganzo, F., O’Donnell, C. J., Gerber, H.-P., & Sapra, P. (2020). PF-06804103, a site-specific anti-HER2 antibody-drug conjugate for the treatment of HER2-expressing breast, gastric, and lung cancers. Molecular Cancer Therapeutics, 19(10), 2068–2078.

Greene, M. K., Chen, T., Robinson, E., Straubinger, N. L., Minx, C., Chan, D. K. W., Wang, J., Burrows, J. F., Van Schaeybroeck, S., Baker, J. R., Caddick, S., Longley, D. B., Mager, D. E., Straubinger, R. M., Chudasama, V. C., & Scott, C. J. (2020). Controlled coupling of an ultrapotent auristatin warhead to cetuximab yields a next-generation antibody-drug conjugate for EGFR-targeted therapy of KRAS mutant pancreatic cancer. British Journal of Cancer, 123(10), 1502–1512.

Hamilton, E. P., Barve, M. B., Bardia, A., Beeram, M., Bendell, J. B., Mosher, R., Hailman, E., Bergstrom, D. A., Burris, H. A., & Soliman, H. H. (2018). Phase 1 dose escalation of XMT-1522, a novel HER2-targeting antibody-drug conjugate (ADC), in patients (pts) with HER2-expressing breast, lung and gastric tumors. Journal of Clinical Oncology, 36(15), 2546–2546.

Hanna, K. S. (2020). Clinical overview of enfortumab vedotin in the management of locally advanced or metastatic urothelial carcinoma. Drugs, 80(1), 1–7.

Hartrampf, N., Seki, T., Baumann, A., Watson, P., Vepřek, N. A., Hetzler, B. E., Hoffmann-Röder, A., Tsuji, M., & Trauner, D. (2020). Optical control of cytokine production using photoswitchable galactosylceramides. Chemistry, 26(20), 4476–4479.

Hasanov, M., Rioth, M. J., Kendra, K., Hernandez-Aya, L., Joseph, R. W., Williamson, S., Chandra, S., Shirai, K., Turner, C. D., Lewis, K., Crowley, E., Moscow, J., Carter, B., & Patel, S. (2020). A phase II study of glembatumumab vedotin for metastatic uveal melanoma. Cancers, 12(8), 2270.

He, C., Hua, X., Sun, S., Li, S., Wang, J., & Huang, X. (2021). Integrated bioinformatic analysis of SARS-CoV-2 infection related genes ACE2, BSG and TMPRSS2 in aerodigestive cancers. Inflammation Researchs, 10(14), 791–802.

Hingorani, D. V., Doan, M. K., Camargo, M. F., Aguilera, J., Song, S. M., Pizzo, D., Scanderbeg, D. J., Cohen, E. E. W., Lowy, A. M., Adams, S. R., & Advani, S. J. (2020). Precision chemoradiotherapy for HER2 tumors using antibody conjugates of an auristatin derivative with reduced cell permeability. Molecular Cancer Therapeutics, 19(1), 157–167.

Hingorani, P., Roth, M. E., Wang, Y., Zhang, W., Gill, J. B., Harrison, D. J., Teicher, B., Erickson, S., Gatto, G., Smith, M. A., Kolb, E. A., & Gorlick, R. (2021). ABBV-085, antibody-drug conjugate targeting LRRC15, is effective in osteosarcoma: A report by the pediatric preclinical testing consortium. Molecular Cancer Therapeutics, 20(3), 535–540.

Hong, D. S., Nicole Concin, N., Vergote, I., De Bono, J., Slomovitz, B. M., Drew, Y., Arkenau, H.-T., Machiels, J.-P., Spicer, J. F., Jones, R., Forster, M. D., Cornez, N., Gennigens, C., Johnson, M., Thistlethwaite, F. C., Rangwala, R. A., Ghatta, S. G., Windfeld, K., Harris, J. R., Lassen, U. N., & Coleman, R. L. (2020). Tisotumab vedotin in previously treated recurrent or metastatic cervical cancer. Clinical Cancer Research, 26(6), 1220–1228.

Hou, X.-M., Hai, Y., Gu, Y.-C., Wang, C.-Y., & Shao, C.-L. (2019). Chemical and bioactive marine natural products of coral-derived microorganisms (2015–2017). Current Medicinal Chemistry, 26(38), 6930–6941.

Hsu, H.-Y., & Hwang, P.-A. (2019). Clinical applications of fucoidan in translational medicine for adjuvant cancer therapy. Clinical and Translational Medicine, 8(1), 15.

Islam, M. M., Shahruzzaman, M., Biswas, S., Sakib, N. M., & Rashid, T. U. (2020). Chitosan based bioactive materials in tissue engineering applications – a review. Bioactive Materials, 5(1), 164–183.

Islam, M. S., Haque, P., Rashid, T. U., Khan, M. N., Mallik, A. M., Khan, M. N. I., & Rahman, M. M. (2017). Core-shell drug carrier from folate conjugated chitosan obtained from prawn shell for targeted doxorubicin delivery. Journal of Materials Science: Materials in Medicine, 28(4), 55.

Jensen, P. R., Moore, B. S., & Fenical, W. (2015). The marine actinomycete genus Salinispora: A model organism for secondary metabolite discovery. Natural Product Reports, 32(5), 738–751.

Jimenez, P. C., Wilke, D. V., Branco, P. C., Bauermeister, A., Rezende-Teixeira, P., Gaudêncio, S. P., & Costa, L. V. (2020). Enriching cancer pharmacology with drugs of marine origin. British Journal of Pharmacology, 177(1), 3–27.

Johansson, M. P., Maaheimo, H., & Ekhol, F. S. (2017). New insight on the structural features of the cytotoxic auristatins MMAE and MMAF revealed by combined NMR spectroscopy and quantum chemical modelling. Scientific Reports, 7, 15920.

Jones, E. B. G., Suetrong, S., & Sakayaroj, J. (2015). Classification of marine Ascomycota, Basidiomycota, Blastocladiomycota and Chytridiomycota. Fungal Diversity, 73, 1–72.

Jones, L., McCalmont, H., Evans, K., Mayoh, C., Kurmasheva, R. T., Billups, C. A., Houghton, P. J., Smith, M. A., & Lock, R. B. (2019). Preclinical activity of the antibody-drug conjugate denintuzumab mafodotin (SGN-CD19A) against pediatric acute lymphoblastic leukemia xenografts. Pediatric Blood and Cancer, 66(8), 27765.

Kadia, T. M., & Gandhi, V. (2017). Nelarabine in the treatment of pediatric and adult patients with T-cell acute lymphoblastic leukemia and lymphoma. Expert Review of Hematology, 10(1), 1–8.

Kalhotra, P., Veera, C. S. R. Chittepu, V. C. S. R., Osorio-Revilla, G., & Gallardo-Velazquez, T. (2021). Field-template, QSAR, ensemble molecular docking, and 3D-RISM solvation studies expose potential of FDA-approved marine drugs as SARS-CoVID-2 main protease inhibitors. Molecules, 26(4), 936.

Kang, H. K., Choi, M. C., & Seo, C. H. (2018). Therapeutic properties and biological benefits of marine-derived anticancer peptides. International Journal of Molecular Sciences, 19(3), 919.

Kaštelan, S., Antunica, A. G., Oresković, L. B., Pelčić, G., Kasun, E., & Hat, K. (2020). Immunotherapy for uveal melanoma – current knowledge and perspectives. Current Medicinal Chemistry, 27(8), 1350–1366.

Katoh, M. (2017). Canonical and non-canonical WNT signaling in cancer stem cells and their niches: Cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (review). International Journal of Oncology, 51(5), 1357–1369.

Kauffmann-Guerrero, D., & Huber, R. M. (2020). Orphan drugs in development for the treatment of small-cell lung cancer: Emerging data on lurbinectedin. Lung Cancer, 11, 27–31.

Kellogg, C. A. (2019). Microbiomes of stony and soft deep-sea corals share rare core bacteria. Microbiome, 7, 90.

Khan, P., Siddiqui, J. A., Lakshmanan, I., Ganti, A. K., Salgia, R., Jain, M., Batra, S. K., & Nasser, M. W. (2021). RNA-based therapies: A cog in the wheel of lung cancer defense. Molecular Cancer, 20, 54.

Khongorzul, P., Ling, C. J., Khan, F. U., Ihsan, A. U. I., & Zhang, J. (2020). Antibody-drug conjugates: A comprehensive review. Molecular Cancer Research, 18(1), 3–19.

Kim, Y. C., & Guan, K. L. (2015). mTOR: A pharmacologic target for autophagy regulation. Journal of Clinical Investigation, 125(1), 25–32.

Knobloch, S., Jóhannsson, R., & Marteinsson, V. (2019). Co-cultivation of the marine sponge Halichondria panicea and its associated microorganisms. Scientific Reports, 9(1), 10403.

Knobloch, S., Jóhannsson, R., & Marteinsson, V. (2020). Genome analysis of sponge symbiont 'Candidatus Halichondribacter symbioticus' shows genomic adaptation to a host-dependent lifestyle. Environmental Microbiology, 22(1), 483–498.

Kobayashi, H., Iwata, S., Wakamatsu, T., Hayakawa, K., Yonemoto, T., Wasa, J., Oka, H., Ueda, T., & Tanaka, S. (2020). Efficacy and safety of trabectedin for patients with unresectable and relapsed soft-tissue sarcoma in Japan: A japanese musculoskeletal oncology group study. Cancer, 126(6), 1253–1263.

Kollmannsberger, C., Choueiri, T. K., Heng, D. Y. C., George, S., Jie, F., Croitoru, R., Poondru, S., & Thompson, J. A. (2021). A randomized phase II study of AGS 16C3F versus axitinib in previously treated patients with metastatic renal cell carcinoma. Oncologist, 26(3), 182–361.

Koopman, L. A., Terp, M. G., Zom, G. Z., Janmaat, M. L., Jacobsen, K., Van den Heuvel, E. G., Brandhorst, M., Forssmann, U., De Bree, F., Pencheva, N., Lingnau, A., Zipeto, M. A., Parren, P. W. H. I., Breij, E. C. W., & Ditzel, H. J. (2019). Enapotamab vedotin, an AXL-specific antibody-drug conjugate, shows preclinical antitumor activity in non-small cell lung cancer. Insight. Journal of Clinical Investigation Insight, 4(21), 128199.

Lammers, R. J., Witjes, W. P., Janzing-Pastors, M. H., Christien, T. M. C., & Witjes, J. A. (2012). Intracutaneous and intravesical immunotherapy with keyhole limpet hemocyanin compared with intravesical mitomycin in patients with non-muscle-invasive bladder cancer: Results from a prospective randomized phase III trial. Journal of Clinical Oncology, 30, 2273–2279.

Le Joncour, V., Martins, A., Puhka, M., Isola, J., Salmikangas, M., Laakkonen, P., Joensuu, H., & Barok, M. (2019). A novel anti-HER2 antibody-drug conjugate XMT-1522 for HER2-positive breast and gastric cancers resistant to trastuzumab emtansine. Molecular Cancer Therapeutics, 18(10), 1721–1730.

Leea, Y., Phatb, C., & Hong, S.-C. (2017). Structural diversity of marine cyclic peptides and their molecular mechanisms for anticancer, antibacterial, antifungal, and other clinical applications. Peptides, 95, 94–105.

Li, L., Xu, M.-Z., Wang, L., Jiang, J., Dong, L.-H., Chen, F., Dong, K., & Song, H.-F. (2020). Conjugating MMAE to a novel anti-HER2 antibody for selective targeted delivery. European Review for Medical and Pharmacological Sciences, 24(24), 12929–12937.

Li, P., Li, L., Wang, S., Liu, Y., Li, Z., & Xia, S. (2021). Effect of antitumor therapy on cancer patients infected by SARS-CoV-2: A systematic review and meta-analysis. Cancer Medicine, 10(5), 1644–1655.

Liersch, T., Meller, J., Kulle, B., Behr, T. M., Markus, P., Langer, C., Ghadimi, B. M., Wegener, W. A., Kovacs, J. K., Horak, I. D., Becker, H., & Goldenberg, D. M. (2005). Phase II trial of carcinoembryonic antigen radioimmunotherapy with 131I-labetuzumab after salvage resection of colorectal metastases in the liver: Five-year safety and efficacy results. Journal of Clinical Oncology, 23(27), 6763–6770.

Lin, Y., Qi, X., Liu, H., Xue, K., Xu, S., & Tian, Z. (2020). The anti-cancer effects of fucoidan: A review of both in vivo and in vitro investigations. Cancer Cell International, 20, 154.

Liu, J. F., Moore, K. N., Birrer, M. J., Berlin, S., Matulonis, U. A., Infante, J. R., Wolpin, B., Poon, K. P., Firestein, R., Xu, J., Kahn, R., Wang, Y., Wood, K., Darbonne, W. C., Lackner, M. R., Kelley, S. K., Lu, X., Choi, Y. J., Maslyar, D., Humke, E. W., & Burris, H. A. (2016). Phase I study of safety and pharmacokinetics of the anti-MUC16 antibody-drug conjugate DMUC5754A in patients with platinum-resistant ovarian cancer or unresectable pancreatic cancer. Annals of Oncology, 27(11), 2124–2130.

Liu, J., Pandya, P., & Afshar, S. (2021). Therapeutic advances in oncology. International Journal of Molecular, 22(4), 2008.

Liu, Z., Frank, M., Yu, X., Yu, H., Tran-Cong, N. M., Gao, Y., & Proksch, P. (2020). Secondary metabolites from marine-derived fungi from China. Progress in the Chemistry of Organic Natural Products, 111, 81–153.

López-Iglesias, A.-A., González-Méndez, L., San-Segundo, L., Herrero, A. B., Hernández-García, S., Martín-Sánchez, M., Gutiérrez, N. C., Paíno, T., Avilés, P., Mateos, M.-V., San-Miguel, J. F., Garayoa, M., & Ocio, E. M. (2017). Synergistic DNA-damaging effect in multiple myeloma with the combination of zalypsis, bortezomib and dexamethasone. Haematologica, 102(1), 168–175.

Losada, A., Berlanga, J. J., Molina-Guijarro, J. M., Jiménez-Ruiz, A., Gago, F., Avilés, P., De Haro, C., & Martínez-Lea, J. F. (2020). Generation of endoplasmic reticulum stress and inhibition of autophagy by plitidepsin induces proteotoxic apoptosis in cancer cells. Biochemical Pharmacology, 172, 113744.

Lowe, K. L., Mackall, C. L., Norry, E., Amado, R., Jakobsen, B. K., & Binder, G. (2018). Fludarabine and neurotoxicity in engineered T-cell therapy. Gene Therapy, 25(3), 176–191.

Luque-Bolivar, A., Pérez-Mora, E., Villegas, V. E., & Rondón-Lagos, M. (2020). Resistance and overcoming resistance in breast cancer. Breast Cancer, 12, 211–229.

Ma, H. L., Xu, Y. F., Qi, X. R., Maitani, Y., & Nagai, T. (2008). Superpara magnetic iron oxide nanoparticles stabilized by alginate: Pharmacokinetics, tissue distribution, and applications in detecting liver cancers. International Journal of Pharmaceutics, 354, 217–226.

Maderna, A., Doroski, M., Subramanyam, C., Porte, A., Leverett, C. A., Vetelino, B. C., Chen, Z., Risley, H., Parris, K., Pandit, J., Varghese, A. H., Shanker, S., Song, C., Sukuru, S. C. K., Farley, K. A., Wagenaar, M. M., Shapiro, M. J., Musto, S., Lam, M.-H., Loganzo, F., & O'Donnell, C. J. (2014). Discovery of cytotoxic dolastatin 10 analogues with N-terminal modifications. Journal of Medicinal Chemistry, 57(24), 10527–10543.

Manzano, А., & Ocaña, А. (2222). Antibody-вrug сonjugates: А рromising novel therapy for the treatment of ovarian cancer. Cancers, 12(8), 2223.

Marchetti, P., Pavan, B., Simoni, D., Baruchello, R., Rondanin, R., Mischiati, C., Feriotto, G., Ferraro, L., Hsu, L.-C., Lee, R. M., & Dalpiaz, A. (2016). A novel hybrid drug between two potent anti-tubulin agents as a potential prolonged anticancer approach. European Journal of Pharmaceutical Sciences, 91, 50–63.

Markham, A. (2020). Lurbinectedin: First approval. Drugs, 80(13), 1345–1353.

Martínez Andrade, K. A., Lauritano, C., Romano, G., & Ianora, A. (2018). Marine microalgae with anti-cancer properties. Marine Drugs, 16(5), 165.

Martínez-Trufero, J., De Sande-González, L. M., Luna, P., Martin-Broto, J., Álvarez, R., Marquina, G., Diaz-Beveridge, R., Poveda, A., Cano, J. M., Cruz-Jurado, J., Pousa, A. L., Salgado, M. A. V., Valverde-Morales, C. M., Sevilla, I., Martínez-García, J., Rubio-Casadevall, J., De Juan, A., Carrasco, J. A., Moura, D. S., Gurruchaga-Sotes, I., & Gutiérrez, A. (2021). A growth modulation index-based GEISTRA score as a new prognostic tool for trabectedin efficacy in patients with advanced soft tissue sarcomas: A spanish group for sarcoma research (GEIS) retrospective study. Cancers, 13(4), 792.

Martins, A., Vieira, H., Gaspar, H., & Santos, S. (2014). Marketed marine natural products in the pharmaceutical and cosmeceutical industries: Tips for success. Marine Drugs, 12(2), 1066–1101.

Matsunaga, Y., Yamaoka, T., Ohba, M., Miura, S., Masuda, H., Sangai, T., Takimoto, M., Nakamura, S., & Tsurutani, J. (2021). Novel anti-FOLR1 antibody-drug conjugate MORAb-202 in breast cancer and non-small cell lung cancer cells. Antibodies, 10(1), 6.

McCarthy, P. J., Roberts, B. F., Carbonell, A., Roberts, J., Wright, A. E., & Chakrabarti, D. (2019). Marine microbiome as a source of antimalarials. Tropical Medicine and Infectious Disease, 4(3), 103.

McNerney, O. K., & Teachey, D. T. (2021). Xenograft models for pediatric cancer therapies. Faculty Reviews, 10, 11.

Mehbub, M. F., Lei, J., Franco, C., Franco, C., & Zhang, W. (2014). Marine sponge derived natural products between 2001 and 2010: Trends and opportunities for discovery of bioactives. Marine Drugs, 12(8), 4539–4577.

Menck, K., Heinrichs, S., Baden, C., & Bleckmann, A. (2021). The WNT/ROR pathway in cancer: From signaling to therapeutic intervention. Cells, 10(1), 142.

Meric-Bernstam, F., Calvo, E., Moreno, V., Chung, H. C., Park, Y. H. P., Bang, Y.-J., Rosen, L. S., Mita, M. M., Garrido-Laguna, I., Leung, A. C. F., Dube, H., Zhong, W., Chen, X., Dawaher, R., & Curigliano, G. (2020). A phase I dose escalation study evaluating the safety and tolerability of a novel anti-HER2 antibody-drug conjugate (PF-06804103) in patients with HER2-positive solid tumors. Journal of Clinical Oncology, 38(15), 1039.

Miller, J. H., Field, J. J., Kanakkanthara, A., Owen, J. G., Singh, A. J., & Northcote, P. T. (2018). Marine invertebrate natural products that target microtubules. Journal of Natural Products, 81(3), 691–702.

Miwa, S., Yamamoto, N., Hayashi, K., Takeuchi, A., Igarashi, K., & Tsuchiya, H. (2019). Therapeutic targets for bone and soft-tissue sarcomas. International Journal of Molecular Sciences, 20(1), 170.

Miyata, Y., Matsuo, T., Ohba, K., Mitsunari, K., Mukae, Y., Otsubo, A., Harada, J., Matsuda, T., Kondo, T., & Sakai, H. (2020). Present status, limitations and future directions of treatment strategies using fucoidan-based therapies in bladder cancer. Cancers, 12(12), 3776.

Moeller, M., Nietzer, S., & Schupp, P. J. (2019). Neuroactive compounds induce larval settlement in the scleractinian coral Leptastrea purpurea. Scientific Reports, 9(1), 2291.

Moore, K. N., Birrer, M. J., Marsters, J., Wang, Y., Choi, Y., Royer-Joo, S., Lemahieu, V., Armstrong, K., Cordova, J., Samineni, D., Schuth, E., Vaze, A., Maslyar, D., Humke, E. W., Hamilton, E. P., & Liu, J. F. (2020). Phase 1b study of anti-NaPi2b antibody-drug conjugate lifastuzumab vedotin (DNIB0600A) in patients with platinum-sensitive recurrent ovarian cancer. Gynecologic Oncology, 158(3), 631–639.

Morell, M., Novotná, E., Milan, J., Danielisová, P., Büküm, N., & Wsól, V. (2021). Selective inhibition of aldo-keto reductase 1C3: A novel mechanism involved in midostaurin and daunorubicin synergism. Archives of Toxicology, 95(1), 67–78.

Morschhauser, F., Flinn, I. W., Advani, R., Sehn, L. H., Diefenbach, C., Kolibaba, K., Press, O. W., Salles, G., Tilly, H., Chen, A. I., Assouline, S., Cheson, B. D., Dreyling, M., Hagenbeek, A., Zinzani, P. L., Jones, S., Cheng, J., Lu, D., Penuel, E., Hirata, J., Wenger, M., Chu, Y. W., & Sharman, J. (2019). Polatuzumab vedotin or pinatuzumab vedotin plus rituximab in patients with relapsed or refractory non-Hodgkin lymphoma: Final results from a phase 2 randomised study (ROMULUS). Lancet Haematology, 6(5), 254–265.

Mukherjee, A., Waters, A. K., Babic, I., Nurmemmedov, E., Glassy, M. C., Kesari, S., & Yenugonda, V. M. (2019). Antibody drug conjugates: progress, pitfalls, and promises. Human Antibodies. 27(1), 53–62.

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. (2014). Microtubule-depolymerizing agents used in antibody-drug conjugates induce antitumor immunity by stimulation of sendritic cells. Cancer Immunology Research, 2(8), 741–55.

Musaimi, O. A., Shaer, D. A., Albericio, F., & De la Torre, B. G. (2021). 2020 FDA TIDES (peptides and oligonucleotides) harvest. Pharmaceuticals, 14(2), 145.

Negi, B., Kumar, D. S., & Rawat, D. (2017). Marine peptides as anticancer agents: A remedy to mankind by nature. Current Protein and Peptide Science, 18(9), 885–904.

Nejadmoghaddam, M. R., Minai-Tehrani, A., Ghahremanzadeh, R., Mahmoudi, M., Dinarvand, R., & Zarnani, A.-H. (2019). Antibody-drug conjugates: Possibilities and challenges. Avicenna Journal of Medical Biotechnology, 11(1), 3–23.

Newman, D. J. (2016). Developing natural product drugs: Supply problems and how they have been overcome. Pharmacology and Therapeutics, 162, 1–9.

Newman, D. J. (2019). The “utility” of highly toxic marine-sourced compounds. Marine Drugs, 17(6), 324.

Newman, D. J., & Cragg, G. M. (2014). Marine-sourced anti-cancer and cancer pain control agents in clinical and late preclinical development. Marine Drugs, 12(1), 255–278.

Newman, D. J., & Cragg, G. M. (2017). Current status of marine-derived compounds as warheads in anti-tumor drug candidates. Marine Drugs, 15(4), 1–19.

Novoselova, M. V., German, S. V., Abakumova, T. O., Perevoschikov, S. V., Sergeeva, O. S., Nesterchuk, M. V., Efimov, O. I., Petrov, K. S., Chernysheva, V. S., Zatsepin, T. S., & Gorin, D. G. (2021). Multifunctional nanostructured drug delivery carriers for cancer therapy: Multimodal imaging and ultrasound-induced drug release. Colloids Surfaces B Biointerfaces, 200, 111576.

Ocio, E. M., Oriol, A., Bladé, J., Teruel, A. I., Martín, J., De la Rubia, J., Gutiérrez, N. C., Díaz-Pavón, J. R., González, S. M., Coronado, C., Fernández-García, E. M., Gómez, M. S., Fernández-Teruel, C., & Miguel, J. S. (2016). Phase I/II study of weekly PM00104 (zalypsis) in patients with relapsed/refractory multiple myeloma. British Journal of Haematology, 172(4), 625–628.

Ogaki, M. B., Coelho, L. C., Vieira, R., Neto, A. A., Zani, C. L., Alves, T. M. A., Junior, P. A. S., Murta, S. M. F., Barbosa, E. C., Oliveira, J. G., Ceravolo, I. P., Pereira, P. O., Cota, B. B., Viana, R. O., Alves, V. S., & Rosa, L. H. (2020). Cultivable fungi present in deep-sea sediments of Antarctica: Taxonomy, diversity, and bioprospecting of bioactive compounds. Extremophiles, 24(2), 227–238.

Orgueira, A. M., Pérez, L. B. P., Torre, A. M. T., Raíndo, A. P. R., López, M. C. L., Arias, J. A. D., Ferro, R. F. F., Rodríguez, B. A. R., Pérez, M. S. G. P., Ferreiro, M. A. F., Vence, N. A. V., Encinas, M. M. P. E., López, J. L. B. L., Martinelli, G., & Cerchione, C. (2020). FLT3 inhibitors in the treatment of acute myeloid leukemia: Current status and future perspectives. Minerva Medica, 111(5), 427–442.

Ou, J., Liu, K., Jiang, J., Wilson, D. A., Liu, L., Wang, F., Wang, S., Tu, Y., & Peng, F. (2020). Micro/nanomotors toward biomedical applications: The recent progress in biocompatibility. Review. Nano-Micro Small, 16(27), 1906184.

Palanisamy, S. K., Rajendran, N. M., & Marino, A. (2017). Natural products diversity of marine ascidians (Tunicates; Ascidiacea) and successful drugs in clinical development. Natural Products and Bioprospecting, 7(1), 1–111.

Park, Y. H., Ahn, H. K., Kim, J.-Y., Ahn, J. S., Im, Y.-H., Kim, S.-H., Lee, S., Chung, H.-S., & Park, S. J. (2020). First-in-human phase I study of ALT-P7, a HER2-targeting antibody-drug conjugate in patients with HER2-positive advanced breast cancer. Journal of Clinical Oncology, 38(15), 3551.

Patrick, A. O., Pavlick, A. C., Johnson, D. B., Hart, L. L., Infante, J. R., Luke, J. J., Lutzky, J., Rothschild, N. E., Spitler, L. E., Cowey, C. L., Alizadeh, A. R., Salama, A. K., He, Y., Hawthorne, T. R., Bagley, R. G., Zhang, J., Turner, C. D., & Hamid, O. (2019). A phase 2 study of glembatumumab vedotin, an antibody-drug conjugate targeting glycoprotein NMB, in patients with advanced melanoma. The Cancer Journal, 125(7), 1113–1123.

Pereira, D. S., Guevara, C. I., Jin, L., Mbong, N., Verlinsky, A., Hsu, S., Aviña, H., Karki, S., Abad, J. D., Yang, P., Moon, S.-J., Malik, F., Choi, M. Y. M., An, Z., Morrison, K., Challita-Eid, P. M., Doñate, F., Joseph, I. B. J., Kipps, T. J., Dick, J. E., & Stover, D. R. (2015). AGS67E, an anti-CD37 monomethyl auristatin E antibody-drug conjugate as a potential therapeutic for B/T-Cell malignancies and AML: A new role for CD37 in AML. Molecular Cancer Therapeutics, 14(7), 1650–1660.

Pereira, R. B., Evdokimov, N. M., Lefranc, F., Valentão, P., Kornienko, A., Pereira, D. P., Andrade, P. B., & Gomes, N. G. M. (2019). Marine-derived anticancer agents: Clinical benefits, innovative mechanisms, and new targets. Marine Drugs, 17(6), 329.

Perez, E. A., Hillman, D. W., Fishkin, P. A., Krook, J. E., Tan, W. W., Kuriakose, P. A., Alberts, S. R., & Dakhil, S. R. (2005). Phase II trial of dolastatin-10 in patients with advanced breast cancer. Investigational New Drugs, 23(3), 257–261.

Pérez-Pérez, M. J., Priego, E. M., & Bueno, O. (2016). Blocking blood flow to solid tumors by destabilizing tubulin: An approach to targeting tumor growth. Journal of Medicinal Chemistry, 59(19), 8685–8711.

Petek, B. J., & Jones, R. L. (2014). PM00104 (Zalypsis®): A marine derived alkylating agent. Molecules, 19(8), 12328–12335.

Petrova, G. V., Kaprin, A. D, Grecova, O. P., & Starinskij, V. V. (2015) Zlokachestvennye novoobrazovanija v Rossii: Obzor statisticheskoj informacii za 1993–2013 gg. [Malignant neoplasms in Russia: Review of statistical information for 1993–2013]. MNIOI im. P. A. Gercena, Moscow (in Russian).

Petrul, H. M., Schatz, C. A., Kopitz, C. C., Adnane, L., McCabe, T. J., Trail, P., Ha, S., Chang, Y. S., Voznesensky, A., Ranges, G., & Tamburini, P. P. (2012). Therapeutic mechanism and efficacy of the antibody-drug conjugate BAY 79-4620 targeting human carbonic anhydrase 9. Molecular Cancer Therapeutics, 11(2), 340–349.

Petrylak, D. P., Heath, E. I., Sonpavde, G., George, S., Morgans, A. K., Eigl, B. J., Picus, J., Cheng, S. Y., Hotte, S. J., Gartner, E. M., Vincent, M., Chu, R., Anand, B., Morrison, K., Jackson, L., Reyno, L. M., & Yu, E. (2016). Anti-tumor activity, safety and pharmacokinetics (PK) of AGS15E (ASG-15ME) in a phase I dose escalation trial in patients (Pts) with metastatic urothelial cancer (mUC). Journal of Clinical Oncology, 34(15), 4532.

Petrylak, D. P., Vogelzang, N. J., Chatta, K., Fleming, M. T., Smith, D. C., Appleman, L. J., Hussain, A., Modiano, M., Singh, P., Tagawa, S. T., Gore, I., McClay, E. F., Mega, A. E., Sartor, A. O., Somer, B., Wadlow, R., Shore, N. D., Olson, W. C., Stambler, N., DiPippo, V. A., & Israel, R. J. (2020). PSMA ADC monotherapy in patients with progressive metastatic castration-resistant prostate cancer following abiraterone and/or enzalutamide: Efficacy and safety in open-label single-arm phase 2 study. Prostate, 80(1), 99–108.

Petty, R., Anthoney, A., Metges, J. P., Alsina, M., Gonçalves, A., Brown, J., Montagut, C., Gunzer, K., Laus, G., Dios, J. L. I., Miguel-Lillo, B., Bohan, P., & Salazar, R. (2016). Phase Ib/II study of elisidepsin in metastatic or advanced gastroesophageal cancer (IMAGE trial). Cancer Chemotherapy and Pharmacology, 77(4), 819–827.

Pimenoff, V. N., Tous, S., Benavente, Y., Alemany, L., Quint, W., Bosch, F. X., Bravo, I. G., & De Sanjosé, S. (2019). Distinct geographic clustering of oncogenic human papillomaviruses multiple infections in cervical cancers: Results from a worldwide cross-sectional study. International Journal of Cancer, 144(10), 2478–2488.

Pivkin, M. V., Kuznecova, T. A., & Sova, V. V. (2006). Morskie griby i ih metabolity [Marine fungi and their metabolites]. Dal’nauka, Vladivostok (in Russian).

Polu, K. R., & Lowman, H. B. (2014). Probody therapeutics for targeting antibodies to diseased tissue. Expert Opinion on Biological Therapy, 14(8), 1049–1053.

Ponziani, S., Di Vittorio, G., Pitari, G., Cimini, A. C., Ardini, M., Gentile, R., Iacobelli, S., Sala, G., Capone, E., Flavell, D. J., Ippoliti, R., & Giansanti, F. (2020). Antibody-drug conjugates: The new frontier of chemotherapy. International Journal of Molecular Sciences, 21(15), 5510.

Popov, A. M. (2006). Protivoopuholevaja aktivnost’ vtorichnyh metabolitov morskih bespozvonochnyh [Antitumor activity of secondary metabolites of marine invertebrates]. Vestnik DVO RAN, 5, 81–90 (in Russian).

Prota, A. E., Bargsten, K., Diaz, J. F., Marsh, M., Cuevas, C., Liniger, M., Neuhaus, C., Andreu, J., Altmann, K.-H., & Steinmetz, M. O. (2014). A new tubulin-binding site and pharmacophore for microtubule-destabilizing anticancer drugs. Proceedings of the National Academy of Sciences USA, 111(38), 13817–13821.

Purcell, J. W., Tanlimco, S. G., Hickson, J., Fox, M., Sho, M., Durkin, L., Uziel, T., Powers, R., Foster, K., McGonigal, T., Kumar, S., Samayoa, J., Zhang, D., Palma, J. P., Mishra, S., Hollenbaugh, D., Gish, K., Morgan-Lappe, S. E., His, E. D., & Chao, D. T. (2018). LRRC15 is a novel mesenchymal protein and stromal target for antibody-drug conjugates. Cancer Research, 78(14), 4059–4072.

Qamar, H., Hussain, K., Soni, A., Khan, A., Hussain, T., & Chénais, B. (2021). Cyanobacteria as natural therapeutics and pharmaceutical potential: Role in antitumor activity and as nanovectors. Molecules, 26(1), 247.

Raghuvanshi, R., & Bharate, S. B. (2020). Preclinical and clinical studies on bryostatins, a class of marine-derived protein kinase C modulators: A mini-review. Current Topics in Medicinal Chemistry, 20(12), 1124–1135.

Rago, B., Tumey, L. N., Wei, C., Barletta, F., Clark, T., Hansel, S., & Han, X. (2017). Quantitative conjugated payload measurement using enzymatic release of antibody-drug conjugate with cleavable linker. Bioconjugate Chemistry, 28(2), 620–626.

Ratnayake, R., Gunasekera, S. P., Ma, J. J., Dang, L. H., Carney, T. J., Paul, V. J., & Luesch, H. (2020). Dolastatin 15 from a marine cyanobacterium suppresses HIF-1α mediated cancer cell viability and vascularization. ChemBioChem, 21(16), 2356–2366.

Ray, A., Okouneva, T., Manna, T., Miller, H. P., Schmid, S., Arthaud, L., Luduena, R., Jordan, M. A., & Wilson, L. (2007). Mechanism of action of the microtubule-targeted antimitotic depsipeptide tasidotin (formerly ILX651) and its major metabolite tasidotin C-carboxylate. Cancer Research, 67(8), 3767–3776.

Remize, M., Brunel, Y., Silva, J. J., & Berthon, J.-Y., Filaire, E. (2021). Microalgae n-3 PUFAs production and use in food and feed industries. Marine Drugs, 19(2), 113.

Riccio, G., Ruocco, N., Mutalipassi, M., Costantini, M., Zupo, V., Coppola, D., Pascale, D. D., & Lauritano, C. (2020). Ten-year research update review: Antiviral activities from aarine organisms. Biomolecules, 10(7), 1007.

Rinnerthaler, G., Gampenrieder, S. P., & Greil, P. (2019). HER2 directed antibody-drug-conjugates beyond T-DM1 in breast cancer. International Journal of Molecular Sciences, 20(5), 1115.

Roach, T. N. F., Little, M., Arts, M. G. I., Huckeba, J., Haas, H. J., George, E. E., Quinn, R. A., Cobián-Güemes, A. G., Naliboff, D. S., Silveira, C. B., Vermeij, M. J. A., Kelly, L. W., Dorrestein, P. C., & Rohwer, F. (2020). A multiomic analysis of in situ coral-turf algal interactions. Proceedings of the National Academy of Sciences of the United States of America, 117(24), 13588–13595.

Roberts, T. C., Langer, R., & Matthew, J. A. (2020). Advances in oligonucleotide drug delivery. Nature Reviews Drug Discovery, 19(10), 673–694.

Rocha-Lima, C. M., Bayraktar, S., Macintyre, J., Raez, L., Flores, A., Ferrell, A., Rubin, E. H., Poplin, E. A., Tan, A. R., Lucarelli, A., & Zojwalla, N. (2010). A phase 1 trial of e7974 administered on day 1 of a 21-day cycle in patients with advanced solid tumors. Cancer, 118(17), 4262–4270.

Rojas, V., Rivas, L., Cárdenas, C., & Guzmán, F. (2020). Cyanobacteria and eukaryotic microalgae as emerging sources of antibacterial peptides. Molecules, 25(24), 5804.

Román, J. J. M., Campo, M. D. C., Villar, J., Paolini, F., Curzio, G., Venuti, A., Jara, L., Ferreira, J., Murgas, P., Lladser, A., Manubens, A., & Becker, M. I. (2019). Immunotherapeutic potential of mollusk hemocyanins in combination with human vaccine adjuvants in murine models of oral cancer. Journal of Immunology Research, 2, 1–19.

Rosa, G.-P., Tavares, W.-R., Sousa, P. M. C., Pagès A. K., Seca, A. M. L., & Pinto, D. C. G. A. (2019). Seaweed secondary metabolites with beneficial health effects: An overview of successes in in vivo studies and clinical trials. Marine Drugs, 18(1), 8.

Roth, P., Mason, W. P., Richardson, P. G., & Weller, M. (2020). Proteasome inhibition for the treatment of glioblastoma. Expert Opinion on Investigational Drugs, 29(10), 1133–1141.

Ruiz-Torres, V., Encinar, J. A., Herranz-López, M., Pérez-Sánchez, A., Galiano, V., Barrajón-Catalán, E., & Vicente, M. (2017). An updated review on marine anticancer compounds: The use of virtual screening for the discovery of small-molecule cancer drugs. Molecules, 22(7), 1037.

Sahlmann, C.-O., Homayounfar, K., Niessner, M., Dyczkowski, J., Conradi, L.-C., Braulke, F., Meller, B., Beibarth, T., Ghadimi, B. M., Meller, J., Goldenberg, D. M., & Liersch, T. (2017). Repeated adjuvant anti-CEA radioimmunotherapy after resection of colorectal liver metastases: safety, feasibility, and long-term efficacy results of a prospective phase 2 study. Cancer, 123(4), 638–649.

Salehi, B., Selamoglu, Z., Mileski, K. S., Pezzani, R., Redaelli, M., Cho, W. C., Kobarfard, F., Rajabi, S., Martorell, M., Kumar, P., Martins, N, Santra, T. S., & Sharifi-Rad, J. (2019). Liposomal cytarabine as cancer therapy: From chemistry to medicine. Biomolecules, 9(12), 773.

Sang, V. T., Dat, T. T. H., Vinh, L. V., Cuong, L. C. V., Oanh, P. T. T., Ha, H., Kim, Y. H., Anh, H. L. T., & Yang, S. Y. (2019). Coral and coral-associated microorganisms: A prolific source of potential bioactive natural products. Marine Drugs, 17(8), 468.

Sansone, C., Bruno, A., Piscitelli, C., Baci, D., Fontana, A., Brunet, C., Noonan, D. N., & Albini, A. (2021). Natural compounds of marine origin as inducers of immunogenic cell death (ICD): Potential role for cancer interception and therapy. Cells, 10(2), 231.

Santos, J. D. S., Vitorino, I., De la Cruz, M., Díaz, C., Cautain, B., Annang, F., Pérez-Moreno, G., Gonzalez, I., Tormo, J. R., Martin, J., Vicente, M. F., & Lage, O. (2020). Diketopiperazines and other bioactive compounds from bacterial symbionts of marine sponges. Antonie Van Leeuwenhoek, 113(7), 875–887.

Savinkova, A. V., Zhidkova, E. M., Tilova, L. R., Lavrova, M. D., Lylova, E. S., Kuzin, K. A., Portjannikova, A. J., Maksimova, V. P., Holodova, A. V., Vlasova, O. A., Fetisov, T. I., Kirsanov, K. I., Belickij, G. A., Jakubovskaja, M. G., & Lesovaja, E. A. (2018). Varianty i perspektivy pereprofilirovanija lekarstvennyh preparatov dlja ispol’zovanija v terapii onkologicheskih zabolevanij [Options and prospects for repurposing drugs for use in the treatment of oncological diseases]. Sibirskij Onkologicheskij Zhurnal, 17(3), 77–87 (in Russian).

Shah, S. A. A., Akhter, N., Auckloo, B. N., Khan, I., Lu, Y., Wang, K., Wu, B., & Guo, Y.-W. (2017). Structural diversity, biological properties and applications of natural products from cyanobacteria. A review. Marine Drugs, 15(11), 354.

Shapiro, G. I., Vaishampayan, G. I., LoRussio, P., Barton, J., Hua, S., Reich, S. D., Shazer, R., Taylor, C. T., Xuan D., & Borghaei, H. (2017). First-in-human trial of an anti-5T4 antibody-monomethylauristatin conjugate, PF-06263507, in patients with advanced solid tumors. Investigational New Drugs, 35(3), 315–323.

Sheng, X., Yan, X., Wang, L., Shi, Y., Yao, X., Luo, H., Shi, B., Liu, J., He, Z., Yu, G., Ying, J., Han, W., Hu, C., Ling, Y., Chi, Z., Cui, C., Si, L., Fang, J., Zhou, A., & Guo, J. (2021). Open-label, multicenter, phase II study of RC48-ADC, a HER2-targeting antibody-drug conjugate, in patients with locally advanced or metastatic urothelial carcinoma. Clinical Cancer Research, 27(1), 43–51.

Sherman, D. J., & Li, J. (2020). Proteasome inhibitors: Harnessing proteostasis to combat disease. Molecules, 25(3), 671.

Shilabin, A. G., & Hamann, M. T. (2011). In vitro and in vivo evaluation of select kahalalide F analogs with antitumor and antifungal activities. Bioorganic and Medicinal Chemistry, 19(22), 6628–6632.

Singh, A. V., Bandi, M., Raje, N., Richardson, P., Palladino, M. A., Chauhan, D., & Anderson, K. C. (2011). A novel vascular disrupting agent plinabulin triggers JNK-mediated apoptosis and inhibits angiogenesis in multiple myeloma cells. Blood, 117(21), 5692–5700.

Singh, I., Lacko, C. S., Zhao, Z., Schmidt, C. E., & Rinaldi, C. (2020). Preparation and evaluation of microfluidic magnetic alginate microparticles for magnetically templated hydrogels. Journal of Colloid and Interface Science, 561, 647–658.

Skidmore, L., Sakamuri, S., Knudsen, N. A., Hewet, A. G., Milutinovic, S., Barkho, W., Biroc, S. L., Kirtley, J., Marsden, R., Storey, K., Lopez, I., Yu, W., Fang, S.-Y., Yao, S., Gu, Y., & Tian, F. (2020). ARX788, a site-specific anti-HER2 antibody-drug conjugate, demonstrates potent and selective activity in HER2-low and T-DM1-resistant breast and gastric cancers. Molecular Cancer Therapeutics, 19(9), 1833–1843.

Smith, T. E., Pond, C. D., Pierce, E., Harmer, Z. P., Kwan, J., Zachariah, M. M., Harper, M. K., Wyche, T. P., Matainaho, T. K., Bugni, T. S., Barrows, L. R., Ireland, C. M., & Schmidt, E. W. (2018). Accessing chemical diversity from the uncultivated symbionts of small marine animals. Nature Chemical Biology, 14(2), 179–185.

Straus, D. J., Długosz-Danecka, M., Alekseev, S., Illés, Á., Picardi, M., Lech-Maranda, E., Feldman, T., Smolewski, P., Savage, K. J., Bartlett, N. L., Walewski, J., Ramchandren, R., Zinzani, P. L., Hutchings, M., Connors, J. M., Radford, J., Munoz, J., Kim, W. S., Advani, R., Ansell, S. M., Younes, A., Miao, H., Liu, R., Fenton, K., Forero-Torres, A., & Gallamini, A. (2020). Brentuximab vedotin with chemotherapy for stage III/IV classical Hodgkin lymphoma: 3-year update of the ECHELON-1 study. Blood, 135(10), 735–742.

Strickler, J. H., Weekes, C. D., Nemunaitis, J., Ramanathan, R. K., Heist, R. S., Morgensztern, D. M., Angevin, E., Bauer, T. M., Yue, H., Motwani, M., Parikh, A., Reilly, E. B., Afar, D., Naumovski, L., & Kelly, K. (2018). First-in-human phase I, dose-escalation and expansion study of telisotuzumab vedotin, an antibody-drug conjugate targeting c-met, in patients mith advanced solid tumors. Journal of Clinical Oncology, 36(33), 3298–3306.

Stuijvenberga, J., Prokschb, P., & Fritz, G. (2020). Targeting the DNA damage response (DDR) by natural compounds. Bioorganic and Medicinal Chemistry, 28(4), 115279.

Subjakova, V., Oravczova, V., & Hianik, T. (2021). Polymer nanoparticles and nanomotors modified by DNA/RNA aptamers and antibodies in targeted therapy of cancer. Polymers, 13(3), 341.

Summer, K., Browne, J., Liu, L., & Benkendorff, K. (2020). Molluscan compounds provide drug leads for the treatment and prevention of respiratory disease. Marine Drugs, 18(11), 570.

Taglialatela-Scafati, O. (2021). New hopes for drugs against COVID-19 come from the sea. Marine Drugs, 19(2), 104.

Takahashi, R., Mabuchi, S., Kawano, M., Sasano, T., Matsumoto, Y., Kuroda, H., Kozasa, K., Hashimoto, K., Sawada, K., & Kimura, T. (2016). Preclinical investigations of PM01183 (lurbinectedin) as a single agent or in combination with other anticancer agents for clear cell carcinoma of the ovary. Plos One, 11(3), 0151050.

Tan, Y. Y., Yap, P. K., Lim, G. L. X., Mehta, M., Chanc, Y., Ng, S. W., Kapoor, D. N., Negi, P., Anand, K., Singh, S. K., Jha, N. K., Lim, L. C., Madheswaran, T., Satija, S., Gupta, G., Dua, K., & Chellappan, D. K. (2020). Perspectives and advancements in the design of nanomaterials for targeted cancer theranostics. Chemico-Biological Interactions, 329, 109221.

Tannir, N. M., Forero-Torres, A., Ramchandren, R., Pal, S. K., Ansell, S. M., Infante, J. R., De Vos, S., Hamlin, P. A., Kim, S. K., Whiting, N. C., Gartner, E. M., Zhao, B., & Thompson, J. A. (2014). Phase I dose-escalation study of SGN-75 in patients with CD70-positive relapsed/refractory non-Hodgkin lymphoma or metastatic renal cell carcinoma. Investigational New Drugs, 32(6), 1246–1257.

Teplinsky, E., & Herzog, T. J. (2017). The efficacy of trabectedin in treating ovarian cancer. Expert Opinion on Pharmacotherapy, 18(3), 313–323.

Tian, Y., Qiu, X., Wang, C., Zhao, J., Jiang, X, Niu, W., Huang, J., & Zhang, F. (2021). Cancer associates with risk and severe events of COVID-19: A systematic review and meta-analysis. International Journal of Cancer, 148(2), 363–374.

Tjuljandin, S. A., & Zhukov, N. V. (2018). Pravda o rossijskoj onkologii: Problemy i vozmozhnye reshenija [The truth about Russian oncology: Problems and possible solutions]. Rossijskoe Obshhestvo Klinicheskoj Onkologii, Moscow (in Russian).

Tolaney, S. M., Barroso-Sousa, R., Keenan, T., Li, T., Trippa, L., Vaz-Luis, I., Wulf, G., Spring, L., Sinclair, N. F., Andrews, C., Pittenger, J., Richardson, E. T., Dillon, D., Lin, N. U., Overmoyer, B., Partridge, A. H., Van Allen, E., Mittendorf, E. A., Winer, E. P., & Krop, I. E. (2020). Effect of eribulin with or without pembrolizumab on progression-free survival for patients with hormone receptor-positive, ERBB2-negative metastatic breast cancer: A randomized clinical trial. JAMA Oncology, 6(10), 1598–1605.

Tolcher, A. W. (2020). The evolution of antibody-drug conjugates: A positive inflexion point. American Society of Clinical Oncology Educational Book, 40, 127–134.

Tonra, J. R., Lloyd, G. K., Mohanlal, R., & Huang, L. (2020). Plinabulin ameliorates neutropenia induced by multiple chemotherapies through a mechanism distinct from G-CSF therapies. Cancer Chemotherapy and Pharmacology, 85(2), 461–468.

Tsai, H.-L., Tai, C.-J., Huang, C.-W., Chang, F.-R., & Wang, J.-Y. (2017). Efficacy of low-molecular-weight fucoidan as a supplemental therapy in metastatic colorectal cancer patients: A double-blind randomized controlled trial. Marine Drugs, 15(4), 122.

Tzogani, K., Penttilä, K., Lähteenvuo, J., Lapveteläinen, T., Anglada, L., Prieto, C., Garcia-Ochoa, B., Enzmann, H., Gisselbrecht, C., Delgado, J., & Pignatti, F. (2021). Ema review of belantamab mafodotin (blenrep) for the treatment of adult patients with relapsed/refractory multiple myeloma. Oncologist, 26(1), 70–76.

Van Andel, L., Rosing, H., Schellens, J. H. M., & Beijnen, J. H. (2018). Review of chromatographic bioanalytical assays for the quantitative determination of marine-derived drugs for cancer treatment. Marine Drugs, 16(7), 246.

Van Den Bent, M., Eoli, M., Sepulveda, J. M., Smits, M., Walenkamp, A., Frenel, J. S., Franceschi, E., Clement, P. M., Chinot, O., De Vos F., Whenham, N., Sanghera, P., Weller, M., Dubbink, H. J., French, P., Looman, J., Dey, J., Krause, S., Ansell, P., Nuyens, S., Spruyt, M., Brilhante, J., Coens, C., Gorlia, T., & Golfinopoulos, V. (2020). Intellance 2/EORTC 1410 randomized phase II study of Depatux-M alone and with temozolomide vs temozolomide or lomustine in recurrent EGFR amplified glioblastoma. Neuro-Oncology, 22(5), 684–693.

Van Stuijvenberg, J., Proksch, P., & Fritz, G. (2020). Targeting the DNA damage response (DDR) by natural compounds. Bioorganic and Medicinal Chemistry, 28(4), 115279.

Vijayakumar, S., & Menakha, M. (2015). Pharmaceutical applications of cyanobacteria. Journal of Acute Medicine, 5(1), 15–23.

Wang, L., Dong, C., Li, X., Han, W., & Xiulan, S. (2017). Anticancer potential of bioactive peptides from animal sources (review). Oncology Reports, 38(2), 637–651.

Wang, P., Kankala, R. K. K., Chen, B., Long, R., Cai, D., Liu, Y., & Wang, S. (2019). Poly-allylamine hydrochloride and fucoidan-based self-assembled polyelectrolyte complex nanoparticles for cancer therapeutics. Journal of Biomedical Materials Research, 107(2), 339–347.

Wang, Q., Liu, S., Yang, F., Gan, L., Yang, X.,

Published
2021-05-14
How to Cite
Bocharova, E. A., Kopytina, N. I., & SlynkoЕ. Е. (2021). Anti-tumour drugs of marine origin currently at various stages of clinical trials (review) . Regulatory Mechanisms in Biosystems, 12(2), 265-280. https://doi.org/10.15421/022136