Depsipeptides Targeting Tumor Cells: Milestones from In Vitro to Clinical Trials
Abstract
:1. Introduction
Methodology Used in Literature Research
2. Depsipeptides with a Recognized Mechanism of Targeting Cancer Cells
2.1. Depsipeptides Inducing Apoptosis in Tumor Cells
2.1.1. Apratoxin A
2.1.2. Aurilides
2.1.3. Beauvericins
2.1.4. Coibamide A
2.1.5. Dehydrodidemnin B
2.1.6. Enniatins
2.1.7. Grassypeptolides
2.1.8. Hantupeptins
2.1.9. Lagunamides
2.1.10. Tiahuramides
2.2. Depsipeptides Inducing Autophagy in Tumor Cells
Beauvenniatins
2.3. Depsipeptides Inhibiting Elastases
2.3.1. Micropeptins
2.3.2. Tutuilamides A–C
2.4. Depsipeptides Inhibiting Histone Deacetylases
2.4.1. Bassianolide
2.4.2. Clavatustides A and B
2.4.3. Cryptophycin
2.4.4. Largazole
2.4.5. Lyngbyabellins
2.4.6. Romidepsin
2.4.7. Sansalvamide A
2.5. Depsipeptides Disrupting Microfilaments
2.5.1. Desmethoxymajusculamide C
2.5.2. Dolastatin 10
2.5.3. Miuraenamide
2.5.4. Nobilamide I
2.6. Depsipeptides Inhibiting Cell Growth
2.6.1. Skyllamycins
2.6.2. Stereocalpin A
2.7. Depsipeptides Inhibiting Topoisomerases
2.7.1. Fusaristatin A
2.7.2. N-Methylsansalvamide
2.8. Oncosis
Kahalalide
3. Depsipeptides Studied In Vivo and Clinical Trials
3.1. Apratoxin
3.2. Beauvericin
3.3. Cryptophycins
3.4. Dolastatin 10
3.5. Largazole
3.6. Plitidepsin (Dehydrodidemnin B)
3.7. Soblidotin
3.8. Tasidotin
4. Current Methods for Purification and Synthesis of Depsipeptides
4.1. Proved Methods for Purifying Depsipeptides
4.2. Synthesis of Depsipeptides
5. Recognized Disadvantages of Depsipeptides
6. Discussion
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Bray, F.; Laversanne, M.; Weiderpass, E.; Soerjomataram, I. The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer 2021, 127, 3029–3030. [Google Scholar] [CrossRef] [PubMed]
- World Health Organization. Global Health Estimates for 2020: Deaths by Cause, Age, Sex, by Country and by Region, 2000–2019; WHO: Geneva, Switzerland, 2020. [Google Scholar]
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA. Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Ferlay, J.; Colombet, M.; Soerjomataram, I.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Cancer statistics for the year 2020: An overview. Int. J. Cancer 2021, 149, 778–789. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
- Pilleron, S.; Sarfati, D.; Janssen-Heijnen, M.; Vignat, J.; Ferlay, J.; Bray, F.; Soerjomataram, I. Global cancer incidence in older adults, 2012 and 2035: A population-based study. Int. J. Cancer 2019, 144, 49–58. [Google Scholar] [CrossRef] [Green Version]
- Pilleron, S.; Soto-Perez-de-Celis, E.; Vignat, J.; Ferlay, J.; Soerjomataram, I.; Bray, F.; Sarfati, D. Estimated global cancer incidence in the oldest adults in 2018 and projections to 2050. Int. J. Cancer 2021, 148, 601–608. [Google Scholar] [CrossRef]
- Trinidad-Calderón, P.A.; López-Castillo, L.-M.; Gallegos-Martínez, S.; Trujillo-de-Santiago, G.; García-Lara, S.; Álvarez, M.M. nurP28, a New-to-Nature Zein-Derived Peptide, Enhances the Therapeutic Effect of Docetaxel in Breast Cancer Monolayers and Spheroids. Molecules 2022, 27, 2824. [Google Scholar] [CrossRef]
- Serna-Thome, G.; Castro-Eguiluz, D.; Fuchs-Tarlovsky, V.; Sanchez-Lopez, M.; Delgado-Olivares, L.; Coronel-Martinez, J.; Molina-Trinidad, E.M.; De La Torre, M.; Cetina-Perez, L. Use of functional foods and oral supplements as adjuvants in cancer treatment. Rev. Investig. Clin. 2018, 70, 136–146. [Google Scholar] [CrossRef] [Green Version]
- Mondal, A.; Bose, S.; Banerjee, S.; Patra, J.K.; Malik, J.; Mandal, S.K.; Kilpatrick, K.L.; Das, G.; Kerry, R.G.; Fimognari, C.; et al. Marine Cyanobacteria and Microalgae Metabolites—A Rich Source of Potential Anticancer Drugs. Mar. Drugs 2020, 18, 476. [Google Scholar] [CrossRef]
- Sharma, P.; Kaur, H.; Kehinde, B.A.; Chhikara, N.; Sharma, D.; Panghal, A. Food-Derived Anticancer Peptides: A Review. Int. J. Pept. Res. Ther. 2021, 27, 55–70. [Google Scholar] [CrossRef]
- Zhang, Q.T.; Liu, Z.D.; Wang, Z.; Wang, T.; Wang, N.; Wang, N.; Zhang, B.; Zhao, Y.F. Recent advances in small peptides of marine origin in cancer therapy. Mar. Drugs 2021, 19, 115. [Google Scholar] [CrossRef]
- Zhang, J.N.; Xia, Y.X.; Zhang, H.J. Natural cyclopeptides as anticancer agents in the last 20 years. Int. J. Mol. Sci. 2021, 22, 3973. [Google Scholar] [CrossRef]
- Adrover-Castellano, M.L.; Schmidt, J.J.; Sherman, D.H. Biosynthetic Cyclization Catalysts for the Assembly of Peptide and Polyketide Natural Products. ChemCatChem 2021, 13, 2095–2116. [Google Scholar] [CrossRef]
- Taevernier, L.; Wynendaele, E.; Gevaert, B.; Spiegeleer, B. Chemical Classification of Cyclic Depsipeptides. Curr. Protein Pept. Sci. 2017, 18, 425–452. [Google Scholar] [CrossRef]
- Alonzo, D.A.; Schmeing, T.M. Biosynthesis of depsipeptides, or Depsi: The peptides with varied generations. Protein Sci. 2020, 29, 2316–2347. [Google Scholar] [CrossRef]
- Rangel, M.; Santana, C.; Pinheiro, A.; Anjos, L.; Barth, T.; Júnior, O.; Fontes, W.; Castro, M. Marine Depsipeptides as Promising Pharmacotherapeutic Agents. Curr. Protein Pept. Sci. 2016, 18, 72–91. [Google Scholar] [CrossRef]
- Lee, Y.; Phat, C.; Hong, S.C. Structural diversity of marine cyclic peptides and their molecular mechanisms for anticancer, antibacterial, antifungal, and other clinical applications. Peptides 2017, 95, 94–105. [Google Scholar] [CrossRef]
- Pavlicevic, M.; Maestri, E.; Marmiroli, M. Marine Bioactive Peptides—An Overview of Generation, Structure and Application with a Focus on Food Sources. Mar. Drugs 2020, 18, 424. [Google Scholar] [CrossRef]
- Khalifa, S.A.M.; Elias, N.; Farag, M.A.; Chen, L.; Saeed, A.; Hegazy, M.E.F.; Moustafa, M.S.; El-Wahed, A.A.; Al-Mousawi, S.M.; Musharraf, S.G.; et al. Marine natural products: A source of novel anticancer drugs. Mar. Drugs 2019, 17, 491. [Google Scholar] [CrossRef] [Green Version]
- Kang, H.; Choi, M.-C.; Seo, C.; Park, Y. Therapeutic Properties and Biological Benefits of Marine-Derived Anticancer Peptides. Int. J. Mol. Sci. 2018, 19, 919. [Google Scholar] [CrossRef]
- D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef] [PubMed]
- Urbańska, K.; Orzechowski, A. Unappreciated role of LDHA and LDHB to control apoptosis and autophagy in tumor cells. Int. J. Mol. Sci. 2019, 20, 2085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pistritto, G.; Trisciuoglio, D.; Ceci, C.; Garufi, A.; D’Orazi, G. Apoptosis as anticancer mechanism: Function and dysfunction of its modulators and targeted therapeutic strategies. Aging 2016, 8, 603–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giampazolias, E.; Tait, S.W.G. Mitochondria and the hallmarks of cancer. FEBS J. 2016, 283, 803–814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, A.; Boise, L.H.; Shanmugam, M. Cancer metabolism and the evasion of apoptotic cell death. Cancers 2019, 11, 1144. [Google Scholar] [CrossRef] [Green Version]
- Moschos, M.M.; Dettoraki, M.; Androudi, S.; Kalogeropoulos, D.; Lavaris, A.; Garmpis, N.; Damaskos, C.; Garmpi, A.; Tsatsos, M. The role of histone deacetylase inhibitors in uveal melanoma: Current evidence. Anticancer Res. 2018, 38, 3817–3824. [Google Scholar] [CrossRef]
- Lee, H.S.; Phat, C.; Choi, S.U.; Lee, C. Synergistic effect of a novel cyclic pentadepsipeptide, neoN-methylsansalvamide, and paclitaxel on human multidrug resistance cancer cell lines. Anticancer Drugs 2013, 24, 455–460. [Google Scholar] [CrossRef]
- Wan, X.; Serrill, J.D.; Humphreys, I.R.; Tan, M.; McPhail, K.L.; Ganley, I.G.; Ishmael, J.E. ATG5 promotes death signaling in response to the cyclic depsipeptides coibamide a and apratoxin A. Mar. Drugs 2018, 16, 77. [Google Scholar] [CrossRef] [Green Version]
- Okada, K.; Hakata, S.; Terashima, J.; Gamou, T.; Habano, W.; Ozawa, S. Combination of the histone deacetylase inhibitor depsipeptide and 5-fluorouracil upregulates major histocompatibility complex class II and p21 genes and activates caspase-3/7 in human colon cancer HCT-116 cells. Oncol. Rep. 2016, 36, 1875–1885. [Google Scholar] [CrossRef] [Green Version]
- Cai, W.; Ratnayake, R.; Gerber, M.H.; Chen, Q.-Y.; Yu, Y.; Derendorf, H.; Trevino, J.G.; Luesch, H. Development of apratoxin S10 (Apra S10) as an anti-pancreatic cancer agent and its preliminary evaluation in an orthotopic patient-derived xenograft (PDX) model. Investig. New Drugs 2019, 37, 364–374. [Google Scholar] [CrossRef]
- Caloni, F.; Fossati, P.; Anadón, A.; Bertero, A. Beauvericin: The beauty and the beast. Environ. Toxicol. Pharmacol. 2020, 75, 103349. [Google Scholar] [CrossRef]
- He, W.; Qiu, H.-B.; Chen, Y.-J.; Xi, J.; Yao, Z.-J. Total synthesis of proposed structure of coibamide A, a highly N- and O-methylated cytotoxic marine cyclodepsipeptide. Tetrahedron Lett. 2014, 55, 6109–6112. [Google Scholar] [CrossRef]
- Zuo, W.; Kwok, H.F. Development of marine-derived compounds for cancer therapy. Mar. Drugs 2021, 19, 342. [Google Scholar] [CrossRef]
- Taevernier, L.; Veryser, L.; Roche, N.; Peremans, K.; Burvenich, C.; Delesalle, C.; De Spiegeleer, B. Human skin permeation of emerging mycotoxins (beauvericin and enniatins). J. Expo. Sci. Environ. Epidemiol. 2016, 26, 277–287. [Google Scholar] [CrossRef]
- Kwan, J.C.; Rocca, J.R.; Abboud, K.A.; Paul, V.J.; Luesch, H. Total structure determination of grassypetolide, a new marine cyanobacterical cytotoxin. Org. Lett. 2008, 10, 789–792. [Google Scholar] [CrossRef]
- Xue, Y.; Zhao, P.; Quan, C.; Zhao, Z.; Gao, W.; Li, J.; Zu, X.; Fu, D.; Feng, S.; Bai, X.; et al. Cyanobacteria-derived peptide antibiotics discovered since 2000. Peptides 2018, 107, 17–24. [Google Scholar] [CrossRef]
- Huang, W.; Ren, R.G.; Dong, H.Q.; Wei, B.G.; Lin, G.Q. Diverse synthesis of marine cyclic depsipeptide lagunamide A and its analogues. J. Org. Chem. 2013, 78, 10747–10762. [Google Scholar] [CrossRef]
- Levert, A.; Alvariño, R.; Bornancin, L.; Abou Mansour, E.; Burja, A.M.; Genevière, A.M.; Bonnard, I.; Alonso, E.; Botana, L.; Banaigs, B. Structures and Activities of Tiahuramides A-C, Cyclic Depsipeptides from a Tahitian Collection of the Marine Cyanobacterium Lyngbya majuscula. J. Nat. Prod. 2018, 81, 1301–1310. [Google Scholar] [CrossRef]
- Qiu, B.; Tan, A.; Veluchamy, A.B.; Li, Y.; Murray, H.; Cheng, W.; Liu, C.; Busoy, J.M.; Chen, Q.-Y.; Sistla, S.; et al. Apratoxin S4 Inspired by a Marine Natural Product, a New Treatment Option for Ocular Angiogenic Diseases. Investig. Opthalmol. Vis. Sci. 2019, 60, 3254. [Google Scholar] [CrossRef] [Green Version]
- Andler, O.; Kazmaier, U. Total synthesis of apratoxin A and B using Matteson’s homologation approach. Org. Biomol. Chem. 2021, 19, 4866–4870. [Google Scholar] [CrossRef]
- Matthew, S.; Schupp, P.J.; Luesch, H. Correction to Apratoxin E, a Cytotoxic Peptolide from a Guamanian Collection of the Marine Cyanobacterium Lyngbya bouillonii. J. Nat. Prod. 2018, 81, 217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eghtedari, M.; Jafari Porzani, S.; Nowruzi, B. Anticancer potential of natural peptides from terrestrial and marine environments: A review. Phytochem. Lett. 2021, 42, 87–103. [Google Scholar] [CrossRef]
- Qamar, H.; Hussain, K.; Soni, A.; Khan, A.; Hussain, T.; Chénais, B. Cyanobacteria as Natural Therapeutics and Pharmaceutical Potential: Role in Antitumor Activity and as Nanovectors. Molecules 2021, 26, 247. [Google Scholar] [CrossRef] [PubMed]
- Rastelli, E.J.; Coltart, D.M. Synthesis and biological activity of apratoxin derivatives. Tetrahedron 2018, 74, 2269–2290. [Google Scholar] [CrossRef]
- Cai, W.; Chen, Q.Y.; Dang, L.H.; Luesch, H. Apratoxin S10, a Dual Inhibitor of Angiogenesis and Cancer Cell Growth to Treat Highly Vascularized Tumors. ACS Med. Chem. Lett. 2017, 8, 1007–1012. [Google Scholar] [CrossRef]
- Cortés-Ciriano, I.; Van Westen, G.J.P.; Bouvier, G.; Nilges, M.; Overington, J.P.; Bender, A.; Malliavin, T.E. Improved large-scale prediction of growth inhibition patterns using the NCI60 cancer cell line panel. Bioinformatics 2016, 32, 85–95. [Google Scholar] [CrossRef] [Green Version]
- Diwan, D.; Cheng, L.; Usmani, Z.; Sharma, M.; Holden, N.; Willoughby, N.; Sangwan, N.; Baadhe, R.R.; Liu, C.; Gupta, V.K. Microbial cancer therapeutics: A promising approach. Semin. Cancer Biol. 2021, 86, 931–950. [Google Scholar] [CrossRef]
- Takase, S.; Kurokawa, R.; Arai, D.; Kanto, K.K.; Okino, T.; Nakao, Y.; Kushiro, T.; Yoshida, M.; Matsumoto, K. A quantitative shRNA screen identifies ATP1A1 as a gene that regulates cytotoxicity by aurilide B. Sci. Rep. 2017, 7, 2002. [Google Scholar] [CrossRef] [Green Version]
- Quintana-Cabrera, R.; Quirin, C.; Glytsou, C.; Corrado, M.; Urbani, A.; Pellattiero, A.; Calvo, E.; Vázquez, J.; Enríquez, J.A.; Gerle, C.; et al. The cristae modulator Optic atrophy 1 requires mitochondrial ATP synthase oligomers to safeguard mitochondrial function. Nat. Commun. 2018, 9, 3399. [Google Scholar] [CrossRef]
- Michon, S.; Cavelier, F.; Salom-Roig, X.J. Synthesis and Biological Activities of Cyclodepsipeptides of Aurilide Family from Marine Origin. Mar. Drugs 2021, 19, 55. [Google Scholar] [CrossRef]
- Hamill, R.L.; Higgens, C.E.; Boaz, H.E.; Gorman, M. The structure op beauvericin, a new depsipeptide antibiotic toxic to artemia salina. Tetrahedron Lett. 1969, 10, 4255–4258. [Google Scholar] [CrossRef]
- Xu, F.; Butler, R.; May, K.; Rexhepaj, M.; Yu, D.; Zi, J.; Chen, Y.; Liang, Y.; Zeng, J.; Hevel, J.; et al. Modified substrate specificity of a methyltransferase domain by protein insertion into an adenylation domain of the bassianolide synthetase. J. Biol. Eng. 2019, 13, 65. [Google Scholar] [CrossRef] [Green Version]
- Urbaniak, M.; Waskiewicz, A.; Stepien, Ł. Fusarium cyclodepsipeptide mycotoxins: Chemistry, biosynthesis, and occurrence. Toxins 2020, 12, 765. [Google Scholar] [CrossRef]
- Mallebrera, B.; Prosperini, A.; Font, G.; Ruiz, M.J. In vitro mechanisms of Beauvericin toxicity: A review. Food Chem. Toxicol. 2018, 111, 537–545. [Google Scholar] [CrossRef]
- Wu, Q.; Patocka, J.; Nepovimova, E.; Kuca, K. A review on the synthesis and a review on the synthesis and bioactivity aspects of beauvericin, a Fusarium mycotoxin. Front. Pharmacol. 2018, 9, 1338. [Google Scholar] [CrossRef] [Green Version]
- Tranter, D.; Paatero, A.O.; Kawaguchi, S.; Kazemi, S.; Serrill, J.D.; Kellosalo, J.; Vogel, W.K.; Richter, U.; Mattos, D.R.; Wan, X.; et al. Coibamide A Targets Sec61 to Prevent Biogenesis of Secretory and Membrane Proteins. ACS Chem. Biol. 2020, 15, 2125–2136. [Google Scholar] [CrossRef]
- Kazemi, S.; Kawaguchi, S.; Badr, C.E.; Mattos, D.R.; Ruiz-Saenz, A.; Serrill, J.D.; Moasser, M.M.; Dolan, B.P.; Paavilainen, V.O.; Oishi, S.; et al. Targeting of HER/ErbB family proteins using broad spectrum Sec61 inhibitors coibamide A and apratoxin A. Biochem. Pharmacol. 2021, 183, 114317. [Google Scholar] [CrossRef]
- Shi, W.; Lu, D.; Wu, C.; Li, M.; Ding, Z.; Li, Y.; Chen, B.; Lin, X.; Su, W.; Shao, X.; et al. Coibamide A kills cancer cells through inhibiting autophagy. Biochem. Biophys. Res. Commun. 2021, 547, 52–58. [Google Scholar] [CrossRef]
- Wu, C.; Cheng, Z.; Lu, D.; Liu, K.; Cheng, Y.; Wang, P.; Zhou, Y.; Li, M.; Shao, X.; Li, H.; et al. Novel N-Methylated Cyclodepsipeptide Prodrugs for Targeted Cancer Therapy. J. Med. Chem. 2021, 64, 991–1000. [Google Scholar] [CrossRef]
- Ali Seyed, M.; Ayesha, S. Marine-derived pipeline anticancer natural products: A review of their pharmacotherapeutic potential and molecular mechanisms. J. Pharm. Sci. 2021, 7, 203. [Google Scholar] [CrossRef]
- Lee, J.; Currano, J.N.; Carroll, P.J.; Joullié, M.M. Didemnins, tamandarins and related natural products. Nat. Prod. Rep. 2012, 29, 404–424. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Lin, X.; Zheng, L. Targeting Cellular Proapoptotic Agents from Marine Sources. In Handbook of Anticancer Drugs from Marine Origin; Springer International Publishing: Cham, Switzerland, 2015; pp. 529–545. [Google Scholar]
- Kumar, A.; Singh, U.K.; Kini, S.G.; Garg, V.; Agrawal, S.; Tomar, P.K.; Pathak, P.; Chaudhary, A.; Gupta, P.; Malik, A. JNK pathway signaling: A novel and smarter therapeutic targets for various biological diseases. Future Med. Chem. 2015, 7, 2065–2086. [Google Scholar] [CrossRef] [PubMed]
- Yue, J.; López, J.M. Understanding MAPK signaling pathways in apoptosis. Int. J. Mol. Sci. 2020, 21, 2346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martínez-Limón, A.; Joaquin, M.; Caballero, M.; Posas, F.; de Nadal, E. The p38 pathway: From biology to cancer therapy. Int. J. Mol. Sci. 2020, 21, 1913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suárez, Y.; González-Santiago, L.; Zarich, N.; Dávalos, A.; Aranda, J.F.; Alonso, M.A.; Lasunción, M.A.; Rojas, J.M.; Muñoz, A. Plitidepsin cellular binding and Rac1/JNK pathway activation depend on membrane cholesterol content. Mol. Pharmacol. 2006, 70, 025569. [Google Scholar] [CrossRef]
- Nalda-Molina, R.; Valenzuela, B.; Ramon-Lopez, A.; Miguel-Lillo, B.; Soto-Matos, A.; Perez-Ruixo, J.J. Population pharmacokinetics meta-analysis of plitidepsin (Aplidin®) in cancer subjects. Cancer Chemother. Pharmacol. 2009, 64, 97–108. [Google Scholar] [CrossRef]
- Le Tourneau, C.; Faivre, S.; Ciruelos, E.; Domínguez, M.J.; López-Martín, J.A.; Izquierdo, M.A.; Jimeno, J.; Raymond, E. Reports of clinical benefit of plitidepsin (Aplidine), a new marine-derived anticancer agent, in patients with advanced medullary thyroid carcinoma. Am. J. Clin. Oncol. Cancer Clin. Trials 2010, 33, 132–136. [Google Scholar] [CrossRef]
- Jaspars, M.; De Pascale, D.; Andersen, J.H.; Reyes, F.; Crawford, A.D.; Ianora, A. The marine biodiscovery pipeline and ocean medicines of tomorrow. J. Mar. Biol. Assoc. United Kingd. 2016, 96, 151–158. [Google Scholar] [CrossRef] [Green Version]
- Spicka, I.; Ocio, E.M.; Oakervee, H.E.; Greil, R.; Banh, R.H.; Huang, S.-Y.; D’Rozario, J.M.; Dimopoulos, M.A.; Martínez, S.; Extremera, S.; et al. Randomized phase III study (ADMYRE) of plitidepsin in combination with dexamethasone vs. dexamethasone alone in patients with relapsed/refractory multiple myeloma. Ann. Hematol. 2019, 98, 2139–2150. [Google Scholar] [CrossRef] [Green Version]
- Mamur, S.; Yuzbasioglu, D.; Yılmaz, S.; Erikel, E.; Unal, F. Assessment of cytotoxic and genotoxic effects of enniatin-A in vitro. Food Addit. Contam. Part A Chem. Anal. Control. Expo. Risk Assess. 2018, 35, 1633–1644. [Google Scholar] [CrossRef]
- Dornetshuber-Fleiss, R.; Heilos, D.; Mohr, T.; Richter, L.; Süssmuth, R.D.; Zlesak, M.; Novicky, A.; Heffeter, P.; Lemmens-Gruber, R.; Berger, W. The naturally born fusariotoxin enniatin B and sorafenib exert synergistic activity against cervical cancer in vitro and in vivo. Biochem. Pharmacol. 2015, 93, 318–331. [Google Scholar] [CrossRef] [Green Version]
- Dornetshuber, R.; Heffeter, P.; Lemmens-Gruber, R.; Elbling, L.; Marko, D.; Micksche, M.; Berger, W. Oxidative stress and DNA interactions are not involved in enniatin- and beauvericin-mediated apoptosis induction. Mol. Nutr. Food Res. 2009, 53, 1112–1122. [Google Scholar] [CrossRef]
- Prosperini, A.; Berrada, H.; Ruiz, M.J.; Caloni, F.; Coccini, T.; Spicer, L.J.; Perego, M.C.; Lafranconi, A. A review of the mycotoxin enniatin B. Front. Public Health 2017, 5, 304. [Google Scholar] [CrossRef]
- Mastanjević, K.; Lukinac, J.; Jukić, M.; Šarkanj, B.; Krstanović, V.; Mastanjević, K. Multi-(Myco)toxins in malting and brewing by-products. Toxins 2019, 11, 30. [Google Scholar] [CrossRef] [Green Version]
- Ahmad, I.Z.; Parvez, S.; Tabassum, H. Cyanobacterial Peptides with Respect to Anticancer Activity: Structural and Functional Perspective, 1st ed.; Elsevier: Amsterdam, The Netherlands, 2021; Volume 67, ISBN 9780128194836. [Google Scholar]
- Fuentes-Tristan, S.; Parra-Saldivar, R.; Iqbal, H.M.N.; Carrillo-Nieves, D. Bioinspired biomolecules: Mycosporine-like amino acids and scytonemin from Lyngbya sp. with UV-protection potentialities. J. Photochem. Photobiol. B Biol. 2019, 201, 111684. [Google Scholar] [CrossRef]
- Dahiya, S.; Dahiya, R. A comprehensive review of chemistry and pharmacological aspects of natural cyanobacterial azoline-based circular and linear oligopeptides. Eur. J. Med. Chem. 2021, 218, 113406. [Google Scholar] [CrossRef]
- Matthews, J.H.; Liang, X.; Paul, V.J.; Luesch, H. A Complementary Chemical and Genomic Screening Approach for Druggable Targets in the Nrf2 Pathway and Small Molecule Inhibitors to Overcome Cancer Cell Drug Resistance. ACS Chem. Biol. 2018, 13, 1189–1199. [Google Scholar] [CrossRef]
- Sun, W.; Yi, Y.; Xia, G.; Zhao, Y.; Yu, Y.; Li, L.; Hua, C.; He, B.; Yang, B.; Yu, C.; et al. Nrf2-miR-129-3p-mTOR Axis Controls an miRNA Regulatory Network Involved in HDACi-Induced Autophagy. Mol. Ther. 2019, 27, 1039–1050. [Google Scholar] [CrossRef]
- You, L.; Peng, H.; Liu, J.; Cai, M.; Wu, H.; Zhang, Z.; Bai, J.; Yao, Y.; Dong, X.; Yin, X.; et al. Catalpol protects arpe-19 cells against oxidative stress via activation of the keap1/nrf2/are pathway. Cells 2021, 10, 2635. [Google Scholar] [CrossRef]
- Wang, J.; Li, X.; Wu, X.; Wang, Z.; Zhang, C.; Cao, G.; Liu, K.; Yan, T. Uncovering the microbiota in renal cell carcinoma tissue using 16S rRNA gene sequencing. J. Cancer Res. Clin. Oncol. 2021, 147, 481–491. [Google Scholar] [CrossRef]
- Fatino, A.; Weese, C.; Valdez, S.; Jiménez-Somarribas, A.; Rafferty, R.J. Synthetic studies towards lagunamide C: Polyketide assembly investigations. Tetrahedron Lett. 2018, 59, 624–627. [Google Scholar] [CrossRef]
- Thuan, N.H.; An, T.T.; Shrestha, A.; Canh, N.X.; Sohng, J.K.; Dhakal, D. Recent Advances in Exploration and Biotechnological Production of Bioactive Compounds in Three Cyanobacterial Genera: Nostoc, Lyngbya, and Microcystis. Front. Chem. 2019, 7, 604. [Google Scholar] [CrossRef] [PubMed]
- Luo, D.; Putra, M.Y.; Ye, T.; Paul, V.J.; Luesch, H. Isolation, structure elucidation and biological evaluation of lagunamide D: A new cytotoxic macrocyclic depsipeptide from marine cyanobacteria. Mar. Drugs 2019, 17, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bonnard, I.; Bornancin, L.; Dalle, K.; Chinain, M.; Zubia, M.; Banaigs, B.; Roué, M. Assessment of the chemical diversity and potential toxicity of benthic cyanobacterial blooms in the Lagoon of Moorea Island (French Polynesia). J. Mar. Sci. Eng. 2020, 8, 406. [Google Scholar] [CrossRef]
- Xu, J.; Zhang, T.; Yao, J.; Lu, J.; Liu, Z.; Ding, L. Recent advances in chemistry and bioactivity of marine cyanobacteria Moorea species. Eur. J. Med. Chem. 2020, 201, 112473. [Google Scholar] [CrossRef]
- Bialik, S.; Dasari, S.K.; Kimchi, A. Autophagy-dependent cell death—Where, how and why a cell eats itself to death. J. Cell Sci. 2018, 131, jcs215152. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Wang, Y.; Huang, W.; Wei, Y.; Jiang, Z.; Kong, L.; Wu, A.A.; Hu, Z.; Huang, H.; Xu, Q.; et al. Biosynthesis and Chemical Diversification of Verucopeptin Leads to Structural and Functional Versatility. Org. Lett. 2020, 22, 4366–4371. [Google Scholar] [CrossRef]
- Kitagaki, J.; Shi, G.; Miyauchi, S.; Murakami, S.; Yang, Y. Cyclic depsipeptides as potential cancer therapeutics. Anticancer Drugs 2015, 26, 259–271. [Google Scholar] [CrossRef]
- Liu, J.; Zhu, X.; Kim, S.J.; Zhang, W. Antimycin-type depsipeptides: Discovery, biosynthesis, chemical synthesis, and bioactivities. Nat. Prod. Rep. 2016, 33, 1146–1165. [Google Scholar] [CrossRef]
- Urbaniak, M.; Stępień, Ł.; Uhlig, S. Evidence for naturally produced beauvericins containing N-Methyl-Tyrosine in Hypocreales Fungi. Toxins 2019, 11, 182. [Google Scholar] [CrossRef]
- Tian, J.; Han, J.J.; Zhang, X.; He, L.W.; Zhang, Y.J.; Bao, L.; Liu, H.W. New Cyclohexadepsipeptides from an Entomogenous Fungus Fusarium proliferatum and Their Cytotoxicity and Autophagy-Inducing Activity. Chem. Biodivers. 2016, 13, 852–860. [Google Scholar] [CrossRef]
- Bunyapaiboonsri, T.; Vongvilai, P.; Auncharoen, P.; Isaka, M. Cyclohexadepsipeptides from the Filamentous Fungus Acremonium sp. BCC 2629. Helv. Chim. Acta 2012, 95, 963–972. [Google Scholar] [CrossRef]
- Marinaccio, L.; Stefanucci, A.; Scioli, G.; Della Valle, A.; Zengin, G.; Cichelli, A.; Mollica, A. Peptide Human Neutrophil Elastase Inhibitors from Natural Sources: An Overview. Int. J. Mol. Sci. 2022, 23, 2924. [Google Scholar] [CrossRef]
- Al-Awadhi, F.H.; Paul, V.J.; Luesch, H. Structural Diversity and Anticancer Activity of Marine-Derived Elastase Inhibitors: Key Features and Mechanisms Mediating the Antimetastatic Effects in Invasive Breast Cancer. ChemBioChem 2018, 19, 815–825. [Google Scholar] [CrossRef]
- Chen, Q.-Y.; Luo, D.; Seabra, G.M.; Luesch, H. Ahp-Cyclodepsipeptides as tunable inhibitors of human neutrophil elastase and kallikrein 7: Total synthesis of tutuilamide A, serine protease selectivity profile and comparison with lyngbyastatin 7. Bioorg. Med. Chem. 2020, 28, 115756. [Google Scholar] [CrossRef]
- Geada, P.; Gkelis, S.; Teixeira, J.; Vasconcelos, V.; Vicente, A.A.; Fernandes, B. Cyanobacterial Toxins as a High Value-Added Product; Elsevier Ltd.: Amsterdam, The Netherlands, 2017; ISBN 9780081010273. [Google Scholar]
- Favas, R.; Morone, J.; Martins, R.; Vasconcelos, V.; Lopes, G. Cyanobacteria and microalgae bioactive compounds in skin-ageing: Potential to restore extracellular matrix filling and overcome hyperpigmentation. J. Enzyme Inhib. Med. Chem. 2021, 36, 1829–1838. [Google Scholar] [CrossRef]
- Swapna, J.; Kanna, G.R.; Raj, P.P.; Elumalai, S.A.K.S.; Sangeetha, T. Evaluation of Anti-Cervical Cancer Potential of Polypeptides from Blue-Green Algae. Int. J. Sci. Eng. Res. 2017, 8, 849–870. [Google Scholar]
- Bolatkhan, K.; Akmukhanova, N.R.; Zayadan, B.K.; Sadvakasova, A.K.; Sinetova, M.A.; Los, D.A. Isolation and Characterization of Toxic Cyanobacteria from Different Natural Sources. Appl. Biochem. Microbiol. 2017, 53, 754–760. [Google Scholar] [CrossRef]
- Ahmad, S.; Saleem, M.; Riaz, N.; Lee, Y.S.; Diri, R.; Noor, A.; Almasri, D.; Bagalagel, A.; Elsebai, M.F. The Natural Polypeptides as Significant Elastase Inhibitors. Front. Pharmacol. 2020, 11, 688. [Google Scholar] [CrossRef]
- Keller, L.; Canuto, K.M.; Liu, C.; Suzuki, B.M.; Almaliti, J.; Sikandar, A.; Naman, C.B.; Glukhov, E.; Luo, D.; Duggan, B.M.; et al. Tutuilamides A-C: Vinyl-Chloride-Containing Cyclodepsipeptides from Marine Cyanobacteria with Potent Elastase Inhibitory Properties. ACS Chem. Biol. 2020, 15, 751–757. [Google Scholar] [CrossRef]
- Rauf, A.; Khalil, A.A.; Olatunde, A.; Khan, M.; Anwar, S.; Alafnan, A.; Rengasamy, K.R. Diversity, molecular mechanisms and structure-activity relationships of marine protease inhibitors—A review. Pharmacol. Res. 2021, 166, 105521. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Shang, Y.P.; Chen, H.Y.; Li, J. Histone deacetylases function as novel potential therapeutic targets for cancer. Hepatol. Res. 2017, 47, 149–159. [Google Scholar] [CrossRef] [PubMed]
- Garmpis, N.; Damaskos, C.; Garmpi, A.; Dimitroulis, D.; Spartalis, E.; Margonis, G.A.; Schizas, D.; Deskou, I.; Doula, C.; Magkouti, E.; et al. Targeting histone deacetylases in malignant melanoma: A future therapeutic agent or just great expectations? Anticancer Res. 2017, 37, 5355–5362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mun, B.; Park, Y.J.; Sung, G.H.; Lee, Y.; Kim, K.H. Synthesis and antitumor activity of (−)-bassianolide in MDA-MB 231 breast cancer cells through cell cycle arrest. Bioorg. Chem. 2016, 69, 64–70. [Google Scholar] [CrossRef] [PubMed]
- Chettu, S.K.; Madhu, R.B.; Raolji, G.B.; Babu, K.R.; Rao, N.S.K.; Gopalakrishnan, S.; Ismail, A.; Reddy, G.B.; Shafi, S. First total synthesis of cyclodepsipeptides clavatustide A and B and their enantiomers. RSC Adv. 2016, 6, 61555–61565. [Google Scholar] [CrossRef]
- Schmidt, J.J.; Khatri, Y.; Brody, S.I.; Zhu, C.; Pietraszkiewicz, H.; Valeriote, F.A.; Sherman, D.H. A versatile chemoenzymatic synthesis for the discovery of potent cryptophycin analogs. ACS Chem. Biol. 2020, 15, 524–532. [Google Scholar] [CrossRef]
- Poli, G.; Di Fabio, R.; Ferrante, L.; Summa, V.; Botta, M. Largazole Analogues as Histone Deacetylase Inhibitors and Anticancer Agents: An Overview of Structure–Activity Relationships. ChemMedChem 2017, 12, 1917–1926. [Google Scholar] [CrossRef]
- Smolewski, P.; Robak, T. The discovery and development of romidepsin for the treatment of T-cell lymphoma. Expert Opin. Drug Discov. 2017, 12, 859–873. [Google Scholar] [CrossRef]
- Romans-Fuertes, P.; Sondergaard, T.E.; Sandmann, M.I.H.; Wollenberg, R.D.; Nielsen, K.F.; Hansen, F.T.; Giese, H.; Brodersen, D.E.; Sørensen, J.L. Identification of the non-ribosomal peptide synthetase responsible for biosynthesis of the potential anti-cancer drug sansalvamide in FusariumFusarium solani. Curr. Genet. 2016, 62, 799–807. [Google Scholar] [CrossRef]
- Kanaoka, M.; Isogai, A.; Suzuki, A.; Tamura, S.; Murakoshi, S.; Ichinoe, M.; Murakoshi, S. Bassianolide, a New Insecticidal Cyclodepsipeptide from Beauveria bassiana and Verticillium lecanii. Agric. Biol. Chem. 1978, 42, 629–635. [Google Scholar] [CrossRef]
- Liao, W.; Lai, T.; Chen, L.; Fu, J.; Sreenivasan, S.T.; Yu, Z.; Ren, J. Synthesis and Characterization of a Walnut Peptides-Zinc Complex and Its Antiproliferative Activity against Human Breast Carcinoma Cells through the Induction of Apoptosis. J. Agric. Food Chem. 2016, 64, 1509–1519. [Google Scholar] [CrossRef]
- Jiang, W.; Ye, P.; Chen, C.T.A.; Wang, K.; Liu, P.; He, S.; Wu, X.; Gan, L.; Ye, Y.; Wu, B. Two novel hepatocellular carcinoma cycle inhibitory cyclodepsipeptides from a hydrothermal vent crab-associated fungus Aspergillus clavatus C2WU. Mar. Drugs 2013, 11, 4761. [Google Scholar] [CrossRef]
- Ye, P.; Shen, L.; Jiang, W.; Ye, Y.; Chen, C.T.A.; Wu, X.; Wang, K.; Wu, B. Zn-driven discovery of a hydrothermal vent fungal metabolite clavatustide C, and an experimental study of the anti-cancer mechanism of clavatustide B. Mar. Drugs 2014, 12, 3203. [Google Scholar] [CrossRef] [Green Version]
- Chettu, S.K.; Konidena, L.N.S.; Korupolu, R.B.; Kameswara Rao, N.S.; Doddipalla, R.; Gandham, H.B.; Guduru, R. Ring opening of benzoxazinones: An improved and efficient synthesis of clavatustides A & B. Tetrahedron Lett. 2017, 58, 3418–3420. [Google Scholar] [CrossRef]
- Tippelt, A.; Busche, T.; Rückert, C.; Nett, M. Complete Genome Sequence of the Cryptophycin-Producing Cyanobacterium Nostoc sp. Strain ATCC 53789. Microbiol. Resour. Announc. 2020, 9, 4–5. [Google Scholar] [CrossRef] [Green Version]
- Mehta, A.; Soni, V.K.; Shukla, D.; Vishvakarma, N.K. Cyanobacteria: A Potential Source of Anticancer Drugs; Academic Press: Cambridge, MA, USA, 2020; ISBN 9780128193112. [Google Scholar]
- Eren, E.; Watts, N.R.; Sackett, D.L.; Wingfield, P.T. Conformational changes in tubulin upon binding cryptophycin-52 reveal its mechanism of action. J. Biol. Chem. 2021, 297, 101138. [Google Scholar] [CrossRef]
- Ahmed, S.; Khan, H.; Fakhri, S.; Aschner, M.; Cheang, W.S. Therapeutic potential of marine peptides in cervical and ovarian cancers. Mol. Cell. Biochem. 2022, 477, 605–619. [Google Scholar] [CrossRef]
- Ghayur, T.; Hugunin, M.; Talanian, R.V.; Ratnofsky, S.; Quinlan, C.; Emoto, Y.; Pandey, P.; Datta, R.; Huang, Y.; Kharbanda, S.; et al. Proteolytic activation of protein kinase C δ by an ICE/CED 3-like protease induces characteristics of apoptosis. J. Exp. Med. 1996, 184, 2399–2404. [Google Scholar] [CrossRef] [Green Version]
- Tian, X.; Li, Y.; Shen, Y.; Li, Q.; Wang, Q.; Feng, L. Apoptosis and inhibition of proliferation of cancer cells induced by cordycepin (Review). Oncol. Lett. 2015, 10, 595–599. [Google Scholar] [CrossRef] [Green Version]
- Bousquet, M.S.; Ma, J.J.; Ratnayake, R.; Havre, P.A.; Yao, J.; Dang, N.H.; Paul, V.J.; Carney, T.J.; Dang, L.H.; Luesch, H. Multidimensional Screening Platform for Simultaneously Targeting Oncogenic KRAS and Hypoxia-Inducible Factors Pathways in Colorectal Cancer. ACS Chem. Biol. 2016, 11, 1322–1331. [Google Scholar] [CrossRef] [Green Version]
- Qiu, B.; Tan, A.; Tan, Y.Z.; Chen, Q.Y.; Luesch, H.; Wang, X. Largazole inhibits ocular angiogenesis by modulating the expression of vegfr2 and p21. Mar. Drugs 2021, 19, 471. [Google Scholar] [CrossRef] [PubMed]
- Law, M.E.; Corsino, P.E.; Jahn, S.C.; Davis, B.J.; Chen, S.; Patel, B.; Pham, K.; Lu, J.; Sheppard, B.; Nørgaard, P.; et al. Glucocorticoids and histone deacetylase inhibitors cooperate to block the invasiveness of basal-like breast cancer cells through novel mechanisms. Oncogene 2013, 32, 1316–1329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Al-Awadhi, F.H.; Salvador-Reyes, L.A.; Elsadek, L.A.; Ratnayake, R.; Chen, Q.-Y.; Luesch, H. Largazole is a Brain-Penetrant Class I HDAC Inhibitor with Extended Applicability to Glioblastoma and CNS Diseases. ACS Chem. Neurosci. 2020, 11, 1937–1943. [Google Scholar] [CrossRef] [PubMed]
- Nehul, J.N. Antineoplastic activity of the extracts from a Cyanobacterium Lyngbya bipunctata Lemm. Int. Res. J. Plant Sci. 2021, 12, 1–4. [Google Scholar]
- Costa, M.; Costa-Rodrigues, J.; Fernandes, M.H.; Barros, P.; Vasconcelos, V.; Martins, R. Marine cyanobacteria compounds with anticancer properties: A review on the implication of apoptosis. Mar. Drugs 2012, 10, 2181–2207. [Google Scholar] [CrossRef] [Green Version]
- Wang, M.; Zhang, J.; He, S.; Yan, X. A review study on macrolides isolated from cyanobacteria. Mar. Drugs 2017, 15, 126. [Google Scholar] [CrossRef] [Green Version]
- Petrich, A.; Nabhan, C. Use of class I histone deacetylase inhibitor romidepsin in combination regimens. Leuk. Lymphoma 2016, 57, 1755–1765. [Google Scholar] [CrossRef]
- Li, L.H.; Zhang, P.R.; Cai, P.Y.; Li, Z.C. Histone deacetylase inhibitor, Romidepsin (FK228) inhibits endometrial cancer cell growth through augmentation of p53-p21 pathway. Biomed. Pharmacother. 2016, 82, 161–166. [Google Scholar] [CrossRef]
- Belofsky, G.N.; Jensen, P.R.; Fenical, W. Sansalvamide: A new cytotoxic cyclic depsipeptide produced by a marine fungus of the genus Fusarium. Tetrahedron Lett. 1999, 40, 2913–2916. [Google Scholar] [CrossRef]
- Hwang, Y.; Rowley, D.; Rhodes, D.; Gertsch, J.; Fenical, W.; Bushman, F. Mechanism of Inhibition of a Poxvirus Topoisomerase by the Marine Natural Product Sansalvamide A. Mol. Pharmacol. 1999, 55, 1049–1053. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Hamann, M.T. Marine Natural Products in the Discovery and Development of Potential Pancreatic Cancer Therapeutics, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2019; Volume 144, ISBN 9780128171554. [Google Scholar]
- Liu, S.; Gu, W.; Lo, D.; Ding, X.Z.; Ujiki, M.; Adrian, T.E.; Soff, G.A.; Silverman, R.B. N-methylsansalvamide a peptide analogues. Potent new antitumor agents. J. Med. Chem. 2005, 48, 3630–3638. [Google Scholar] [CrossRef]
- Zhang, G.; Liu, S.; Liu, Y.; Wang, F.; Ren, J.; Gu, J.; Zhou, K.; Shan, B. A novel cyclic pentapeptide, H-10, inhibits B16 cancer cell growth and induces cell apoptosis. Oncol. Lett. 2014, 8, 248–252. [Google Scholar] [CrossRef]
- Kigoshi, H.; Kita, M. Antitumor Effects of Sea Hare-Derived Compounds in Cancer. In Handbook of Anticancer Drugs from Marine Origin; Springer International Publishing: Cham, Switzerland, 2015; pp. 701–739. ISBN 9783319071459. [Google Scholar]
- Tan, L.T.; Gupta, D.K. Molecular Targets of Anticancer Agents from Filamentous Marine Cyanobacteria. In Handbook of Anticancer Drugs from Marine Origin; Springer International Publishing: Cham, Switzerland, 2015; pp. 571–592. ISBN 9783319071459. [Google Scholar]
- Mandal, S.; Rath, J. Anticancer Drug Development from Cyanobacteria. In Extremophilic Cyanobacteria for Novel Drug Development. SpringerBriefs in Pharmaceutical Science & Drug Development; Springer: Cham, Switzerland, 2015; pp. 63–78. [Google Scholar] [CrossRef]
- Śliżewska, A.; Żymańczyk-Duda, E. Cyanobacteria as valuable tool in biotechnology. Catalysts 2021, 11, 1259. [Google Scholar] [CrossRef]
- Zhou, W.; Nie, X.D.; Zhang, Y.; Si, C.M.; Zhou, Z.; Sun, X.; Wei, B.G. A practical approach to asymmetric synthesis of dolastatin 10. Org. Biomol. Chem. 2017, 15, 6119–6131. [Google Scholar] [CrossRef] [Green Version]
- Tost, M.; Andler, O.; Kazmaier, U. A Matteson Homologation-Based Synthesis of Doliculide and Derivatives. Eur. J. Org. Chem. 2021, 2021, 6459–6471. [Google Scholar] [CrossRef]
- Yamashita, T.; Matoba, H.; Kuranaga, T.; Inoue, M. Total syntheses of nobilamides B and D: Application of traceless Staudinger ligation. Tetrahedron 2014, 70, 7746–7752. [Google Scholar] [CrossRef]
- Ahmed, S.; Mirzaei, H.; Aschner, M.; Khan, A.; Al-Harrasi, A.; Khan, H. Marine peptides in breast cancer: Therapeutic and mechanistic understanding. Biomed. Pharmacother. 2021, 142, 112038. [Google Scholar] [CrossRef]
- Gao, G.; Wang, Y.; Hua, H.; Li, D.; Tang, C. Marine antitumor peptide dolastatin 10: Biological activity, structural modification and synthetic chemistry. Mar. Drugs 2021, 19, 363. [Google Scholar] [CrossRef]
- Wang, X.; Dong, S.; Feng, D.; Chen, Y.; Ma, M.; Hu, W. Synthesis and biological activity evaluation of dolastatin 10 analogues with N-terminal modifications. Tetrahedron 2017, 73, 2255–2266. [Google Scholar] [CrossRef]
- Fanale, D.; Bronte, G.; Passiglia, F.; Calò, V.; Castiglia, M.; Di Piazza, F.; Barraco, N.; Cangemi, A.; Catarella, M.T.; Insalaco, L.; et al. Stabilizing versus destabilizing the microtubules: A double-edge sword for an effective cancer treatment option? Anal. Cell. Pathol. 2015, 2015, 690916. [Google Scholar] [CrossRef] [Green Version]
- Komal, A. Biologically active peptides from marine proteobacteria: Discussion article. Open J. Bacteriol. 2021, 5, 005–012. [Google Scholar] [CrossRef]
- Wang, S.; Crevenna, A.H.; Ugur, I.; Marion, A.; Antes, I.; Kazmaier, U.; Hoyer, M.; Lamb, D.C.; Gegenfurtner, F.; Kliesmete, Z.; et al. Actin stabilizing compounds show specific biological effects due to their binding mode. Sci. Rep. 2019, 9, 9731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moser, C.; Rüdiger, D.; Förster, F.; Von Blume, J.; Yu, P.; Kuster, B.; Kazmaier, U.; Vollmar, A.M.; Zahler, S. Persistent inhibition of pore-based cell migration by sub-toxic doses of miuraenamide, an actin filament stabilizer. Sci. Rep. 2017, 7, 16407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gemperlein, K.; Zaburannyi, N.; Garcia, R.; La Clair, J.J.; Müller, R. Metabolic and biosynthetic diversity in marine myxobacteria. Mar. Drugs 2018, 16, 314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karuppiah, V.; Sun, W.; Li, Z. Natural Products of Actinobacteria Derived from Marine Organisms. Stud. Nat. Prod. Chem. 2016, 48, 417–446. [Google Scholar] [CrossRef]
- Le, T.C.; Pulat, S.; Lee, J.; Kim, G.J.; Kim, H.; Lee, E.Y.; Hillman, P.F.; Choi, H.; Yang, I.; Oh, D.C.; et al. Marine Depsipeptide Nobilamide i Inhibits Cancer Cell Motility and Tumorigenicity via Suppressing Epithelial-Mesenchymal Transition and MMP2/9 Expression. ACS Omega 2022, 7, 1722–1732. [Google Scholar] [CrossRef]
- Dominguez-Brauer, C.; Thu, K.L.; Mason, J.M.; Blaser, H.; Bray, M.R.; Mak, T.W. Targeting Mitosis in Cancer: Emerging Strategies. Mol. Cell 2015, 60, 524–536. [Google Scholar] [CrossRef] [Green Version]
- Haschka, M.; Karbon, G.; Fava, L.L.; Villunger, A. Perturbing mitosis for anti-cancer therapy: Is cell death the only answer? EMBO Rep. 2018, 19, e45440. [Google Scholar] [CrossRef]
- Zhu, J.; Zhang, S.; Zechel, D.L.; Paululat, T.; Bechthold, A. Rational Design of Hybrid Natural Products by Utilizing the Promiscuity of an Amide Synthetase. ACS Chem. Biol. 2019, 14, 1793–1801. [Google Scholar] [CrossRef]
- Byeon, H.E.; Park, B.K.; Yim, J.H.; Lee, H.K.; Moon, E.Y.; Rhee, D.K.; Pyo, S. Stereocalpin A inhibits the expression of adhesion molecules in activated vascular smooth muscle cells. Int. Immunopharmacol. 2012, 12, 315–325. [Google Scholar] [CrossRef]
- Giltrap, A.M.; Haeckl, F.P.J.; Kurita, K.L.; Linington, R.G.; Payne, R.J. Synthetic Studies Toward the Skyllamycins: Total Synthesis and Generation of Simplified Analogues. J. Org. Chem. 2018, 83, 7250–7270. [Google Scholar] [CrossRef]
- Cimmino, A.; Nimis, P.L.; Masi, M.; De Gara, L.; van Otterlo, W.A.L.; Kiss, R.; Evidente, A.; Lefranc, F. Have lichenized fungi delivered promising anticancer small molecules? Phytochem. Rev. 2019, 18, 1–36. [Google Scholar] [CrossRef]
- Liu, M.; Liu, N.; Shang, F.; Huang, Y. Activation and Identification of NC-1: A Cryptic Cyclodepsipeptide from Red Soil-Derived Streptomyces sp. FXJ1.172. Eur. J. Org. Chem. 2016, 2016, 3943–3948. [Google Scholar] [CrossRef]
- Melander, R.J.; Basak, A.K.; Melander, C. Natural products as inspiration for the development of bacterial antibiofilm agents. Nat. Prod. Rep. 2020, 37, 1454–1477. [Google Scholar] [CrossRef]
- Sweeney, P.; Murphy, C.D.; Caffrey, P. Exploiting the genome sequence of Streptomyces nodosus for enhanced antibiotic production. Appl. Microbiol. Biotechnol. 2016, 100, 1285–1295. [Google Scholar] [CrossRef] [Green Version]
- Pohle, S.; Appelt, C.; Roux, M.; Fiedler, H.P.; Süssmuth, R.D. Biosynthetic gene cluster of the non-ribosomally synthesized cyclodepsipeptide skyllamycin: Deciphering unprecedented ways of unusual hydroxylation reactions. J. Am. Chem. Soc. 2011, 133, 6194–6205. [Google Scholar] [CrossRef]
- Seo, C.; Yim, J.H.; Lee, H.K.; Park, S.M.; Sohn, J.H.; Oh, H. Stereocalpin A, a bioactive cyclic depsipeptide from the Antarctic lichen Stereocaulon alpinum. Tetrahedron Lett. 2008, 49, 29–31. [Google Scholar] [CrossRef]
- Murai, J.; Thomas, A.; Miettinen, M.; Pommier, Y. Schlafen 11 (SLFN11), a restriction factor for replicative stress induced by DNA-targeting anti-cancer therapies. Pharmacol. Ther. 2019, 201, 94–102. [Google Scholar] [CrossRef]
- Mohan, C.D.; Rangappa, S.; Nayak, S.C.; Jadimurthy, R.; Wang, L.; Sethi, G.; Garg, M.; Rangappa, K.S. Bacteria as a treasure house of secondary metabolites with anticancer potential. Semin. Cancer Biol. 2021, 86, 998–1013. [Google Scholar] [CrossRef]
- Watters, D.J. Ascidian toxins with potential for drug development. Mar. Drugs 2018, 16, 162. [Google Scholar] [CrossRef] [Green Version]
- Hegge, A.; Lønborg, R.; Nielsen, D.M.; Sørensen, J.L. Factors influencing production of fusaristatin A in Fusarium graminearum. Metabolites 2015, 5, 184–191. [Google Scholar] [CrossRef] [PubMed]
- Toghueo, R.M.K. Bioprospecting endophytic fungi from Fusarium genus as sources of bioactive metabolites. Mycology 2020, 11, 1–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shiono, Y.; Tsuchinari, M.; Shimanuki, K.; Miyajima, T.; Murayama, T.; Koseki, T.; Laatsch, H.; Funakoshi, T.; Takanami, K.; Suzuki, K. Fusaristatins A and B, two new cyclic lipopeptides from an endophytic Fusarium sp. J. Antibiot. (Tokyo) 2007, 60, 309–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saravanakumar, K.; Rajendren, N.; Kathiresan, K.; Wang, M. Medicinal Drug-related Bioactive Agents from Marine Fungi. Encycl. Mar. Biotechnol. 2020, 4, 2173–2190. [Google Scholar] [CrossRef]
- Cui, H.; Yu, J.; Chen, S.; Ding, M.; Huang, X.; Yuan, J.; She, Z. Alkaloids from the mangrove endophytic fungus Diaporthe phaseolorum SKS019. Bioorganic Med. Chem. Lett. 2017, 27, 803–807. [Google Scholar] [CrossRef]
- Cueto, M.; Jensen, P.R.; Fenical, W. N-Methylsansalvamide, a cytotoxic cyclic depsipeptide from a marine fungus of the genus Fusarium. Phytochemistry 2000, 55, 223–226. [Google Scholar] [CrossRef]
- Song, H.H.; Lee, H.S.; Lee, C. A new cytotoxic cyclic pentadepsipeptide, neo-N-methylsansalvamide produced by FusariumFusarium solani KCCM90040, isolated from potato. Food Chem. 2011, 126, 472–478. [Google Scholar] [CrossRef]
- Puig, B.; Brenna, S.; Magnus, T. Molecular communication of a dying neuron in stroke. Int. J. Mol. Sci. 2018, 19, 2834. [Google Scholar] [CrossRef] [Green Version]
- Weerasinghe, P.; Hallock, S.; Brown, R.; Buja, L.M. Oncosis. In Apoptosis and Beyond; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2018; pp. 567–582. [Google Scholar]
- Tsai, C.M. AqF026 may act as a cancer therapeutic agent via inducing cancer cell oncosis. Med. Hypotheses 2020, 140, 109685. [Google Scholar] [CrossRef]
- Dayanidhi, D.L.; Thomas, B.C.; Osterberg, J.S.; Vuong, M.; Vargas, G.; Kwartler, S.K.; Schmaltz, E.; Dunphy-Daly, M.M.; Schultz, T.F.; Rittschof, D.; et al. Exploring the Diversity of the Marine Environment for New Anti-cancer Compounds. Front. Mar. Sci. 2021, 7, 614766. [Google Scholar] [CrossRef]
- Wyer, S.; Townsend, D.M.; Ye, Z.; Kourtidis, A.; Choo, Y.-M.; de Barros, A.L.B.; Donia, M.S.; Hamann, M.T. Recent advances and limitations in the application of kahalalides for the control of cancer. Biomed. Pharmacother. 2022, 148, 112676. [Google Scholar] [CrossRef] [PubMed]
- Wang, B.; Waters, A.L.; Valeriote, F.A.; Hamann, M.T. An efficient and cost-effective approach to kahalalide F N-terminal modifications using a nuisance algal bloom of Bryopsis pennata. Biochim. Biophys. Acta Gen. Subj. 2015, 1850, 1849–1854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hamann, M.T. Technology evaluation: Kahalalide F, PharmaMar. Curr. Opin. Mol. Ther. 2004, 6, 657–665. [Google Scholar] [PubMed]
- Ciavatta, M.L.; Lefranc, F.; Carbone, M.; Mollo, E.; Gavagnin, M.; Betancourt, T.; Dasari, R.; Kornienko, A.; Kiss, R. Marine Mollusk-Derived Agents with Antiproliferative Activity as Promising Anticancer Agents to Overcome Chemotherapy Resistance. Med. Res. Rev. 2017, 37, 702–801. [Google Scholar] [CrossRef] [PubMed]
- Ha, M.W.; Song, B.R.; Chung, H.J.; Paek, S.M. Design and synthesis of anti-cancer chimera molecules based on marine natural products. Mar. Drugs 2019, 17, 500. [Google Scholar] [CrossRef] [Green Version]
- Rosa, G.P.; Tavares, W.R.; Sousa, P.M.C.; Pagès, A.K.; Seca, A.M.L.; Pinto, D.C.G.A. Seaweed secondary metabolites with beneficial health effects: An overview of successes in in vivo studies and clinical trials. Mar. Drugs 2020, 18, 8. [Google Scholar] [CrossRef] [Green Version]
- Khalifa, S.A.M.; Shedid, E.S.; Saied, E.M.; Jassbi, A.R.; Jamebozorgi, F.H.; Rateb, M.E.; Du, M.; Abdel-Daim, M.M.; Kai, G.Y.; Al-Hammady, M.A.M.; et al. Cyanobacteria—From the oceans to the potential biotechnological and biomedical applications. Mar. Drugs 2021, 19, 241. [Google Scholar] [CrossRef]
- Chen, Q.Y.; Liu, Y.; Luesch, H. Systematic chemical mutagenesis identifies a potent novel apratoxin A/E hybrid with improved in vivo antitumor activity. ACS Med. Chem. Lett. 2011, 2, 861–865. [Google Scholar] [CrossRef]
- Luesch, H.; Yoshida, W.Y.; Moore, R.E.; Paul, V.J.; Corbett, T.H. Total structure determination of apratoxin A, a potent novel cytotoxin from the marine cyanobacterium Lyngbya majuscula. J. Am. Chem. Soc. 2001, 123, 5418–5423. [Google Scholar] [CrossRef]
- Huang, K.C.; Chen, Z.; Jiang, Y.; Akare, S.; Kolber-Simonds, D.; Condon, K.; Agoulnik, S.; Tendyke, K.; Shen, Y.; Wu, K.M.; et al. Apratoxin a shows novel pancreas-targeting activity through the binding of sec 61. Mol. Cancer Ther. 2016, 15, 1208–1216. [Google Scholar] [CrossRef] [Green Version]
- Heilos, D.; Rodríguez-Carrasco, Y.; Englinger, B.; Timelthaler, G.; Van Schoonhoven, S.; Sulyok, M.; Boecker, S.; Süssmuth, R.D.; Heffeter, P.; Lemmens-Gruber, R.; et al. The natural fungal metabolite beauvericin exerts anticancer activity in vivo: A pre-clinical pilot study. Toxins 2017, 9, 258. [Google Scholar] [CrossRef]
- Cazzamalli, S.; Figueras, E.; Petho, L.; Borbély, A.; Steinkühler, C.; Neri, D.; Sewald, N. In Vivo Antitumor Activity of a Novel Acetazolamide-Cryptophycin Conjugate for the Treatment of Renal Cell Carcinomas. ACS Omega 2018, 3, 14726–14731. [Google Scholar] [CrossRef]
- Weiss, C.; Figueras, E.; Borbely, A.N.; Sewald, N. Cryptophycins: Cytotoxic cyclodepsipeptides with potential for tumor targeting. J. Pept. Sci. 2017, 23, 514–531. [Google Scholar] [CrossRef] [Green Version]
- Nowruzi, B.; Haghighat, S.; Fahimi, H.; Mohammadi, E. Nostoc cyanobacteria species: A new and rich source of novel bioactive compounds with pharmaceutical potential. J. Pharm. Health Serv. Res. 2018, 9, 5–12. [Google Scholar] [CrossRef]
- Mohammad, R.M.; Pettit, G.R.; Almatchy, V.P.; Wall, N.; Varterasian, M.; AI-Katib, A. Synergistic interaction of selected marine animal anticancer drugs against human diffuse large cell lymphoma. Anticancer Drugs 1998, 9, 149–156. [Google Scholar] [CrossRef]
- Kalemkerian, G.P.; Ou, X.; Adil, M.R.; Rosati, R.; Khoulani, M.M.; Madan, S.K.; Pettit, G.R. Activity of dolastatin 10 against small-cell lung cancer in vitro and in vivo: Induction of apoptosis and bcl-2 modification. Cancer Chemother. Pharmacol. 1999, 43, 507–515. [Google Scholar] [CrossRef]
- Beesoo, R.; Neergheen-Bhujun, V.; Bhagooli, R.; Bahorun, T. Apoptosis inducing lead compounds isolated from marine organisms of potential relevance in cancer treatment. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2014, 768, 84–97. [Google Scholar] [CrossRef]
- Wang, X.; Yu, L.; Liu, Z.; Xu, P.; Tan, H.; Wu, T.; Zeng, W. Advances of Microtubule-Targeting Small Molecular Anticancer Agents from Marine Origin. In Handbook of Anticancer Drugs from Marine Origin; Springer International Publishing: Cham, Switzerland, 2015; pp. 481–513. [Google Scholar]
- Hong, J.; Luesch, H. Largazole: From discovery to broad-spectrum therapy. Nat. Prod. Rep. 2012, 29, 449–456. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Salvador, L.A.; Byeon, S.; Ying, Y.; Kwan, J.C.; Law, B.K.; Hong, J.; Luesch, H. Anticolon cancer activity of largazole, a marine-derived tunable histone deacetylase inhibitor. J. Pharmacol. Exp. Ther. 2010, 335, 351–361. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Wang, Z.; Wang, J.; Lam, W.; Kwong, S.; Li, F.; Friedman, S.L.; Zhou, S.; Ren, Q.; Xu, Z.; et al. A histone deacetylase inhibitor, largazole, decreases liver fibrosis and angiogenesis by inhibiting transforming growth factor-β and vascular endothelial growth factor signalling. Liver Int. 2013, 33, 504–515. [Google Scholar] [CrossRef]
- Pilon, J.L.; Clausen, D.J.; Hansen, R.J.; Lunghofer, P.J.; Charles, B.; Rose, B.J.; Thamm, D.H.; Gustafson, D.L.; Bradner, J.E.; Williams, R.M. Comparative pharmacokinetic properties and antitumor activity of the marine HDACi Largazole and Largazole peptide isostere. Cancer Chemother. Pharmacol. 2015, 75, 671–682. [Google Scholar] [CrossRef] [PubMed]
- Delgado-Calle, J.; Kurihara, N.; Atkinson, E.G.; Nelson, J.; Miyagawa, K.; Galmarini, C.M.; Roodman, G.D.; Bellido, T. Aplidin (plitidepsin) is a novel anti-myeloma agent with potent anti-resorptive activity mediated by direct effects on osteoclasts. Oncotarget 2019, 10, 2709–2721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cooper, E.L.; Albert, R. Tunicates: A Vertebrate Ancestral Source of Antitumor Compounds. In Handbook of Anticancer Drugs from Marine Origin; Springer International Publishing: Cham, Switzerland, 2015; pp. 383–395. [Google Scholar]
- Lemmens-Gruber, R. Antitumour Effect of Cyclodepsipeptides from Marine Sponges. In Handbook of Anticancer Drugs from Marine Origin; Springer International Publishing: Cham, Switzerland, 2015; pp. 101–111. [Google Scholar]
- Caers, J.; Menu, E.; De Raeve, H.; Lepage, D.; Van Valckenborgh, E.; Van Camp, B.; Alvarez, E.; Vanderkerken, K. Antitumour and antiangiogenic effects of Aplidin® in the 5TMM syngeneic models of multiple myeloma. Br. J. Cancer 2008, 98, 1966–1974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barboza, N.M.; Medina, D.J.; Budak-Alpdogan, T.; Aracil, M.; Jimeno, J.M.; Bertino, J.R.; Banerjee, D. Plitidepsin (Aplidin) is a potent inhibitor of diffuse large cell and Burkitt lymphoma and is synergistic with rituximab. Cancer Biol. Ther. 2012, 13, 114–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muñoz-Alonso, M.J.; González-Santiago, L.; Martínez, T.; Losada, A.; Galmarini, C.M.; Muñoz, A. The mechanism of action of plitidepsin. Curr. Opin. Investig. Drugs 2009, 10, 536–542. [Google Scholar]
- Ribrag, V.; Caballero, D.; Fermé, C.; Zucca, E.; Arranz, R.; Briones, J.; Gisselbrecht, C.; Salles, G.; Gianni, A.M.; Gomez, H.; et al. Multicenter phase II study of plitidepsin in patients with relapsed/refractory non-Hodgkin’s lymphoma. Haematologica 2013, 98, 357. [Google Scholar] [CrossRef]
- Florean, C.; Dicato, M.; Diederich, M. Immune-modulating and anti-inflammatory marine compounds against cancer. Semin. Cancer Biol. 2022, 80, 58–72. [Google Scholar] [CrossRef]
- Alonso-Álvarez, S.; Pardal, E.; Sánchez-Nieto, D.; Navarro, M.; Caballero, M.D.; Mateos, M.V.; Martín, A. Plitidepsin: Design, development, and potential place in therapy. Drug Des. Dev. Ther. 2017, 11, 253. [Google Scholar] [CrossRef] [Green Version]
- Wang, E.; Sorolla, M.A.; Krishnan, P.D.G.; Sorolla, A. From seabed to bedside: A review on promising marine anticancer compounds. Biomolecules 2020, 10, 248. [Google Scholar] [CrossRef] [Green Version]
- Watanabe, J.; Minami, M.; Kobayashi, M. Antitumor activity of TZT-1027 (soblidotin). Anticancer Res. 2006, 26, 1973–1981. [Google Scholar]
- Natsume, T.; Watanabe, J.I.; Koh, Y.; Fujio, N.; Ohe, Y.; Horiuchi, T.; Saijo, N.; Nishio, K.; Kobayashi, M. Antitumor activity of TZT-1027 (Soblidotin) against vascular endothelial growth factor-secreting human lung cancer in vivo. Cancer Sci. 2003, 94, 826–833. [Google Scholar] [CrossRef] [Green Version]
- Dahiya, R.; Dahiya, S.; Fuloria, N.K.; Kumar, S.; Mourya, R.; Chennupati, S.V.; Jankie, S.; Gautam, H.; Singh, S.; Karan, S.K.; et al. Natural bioactive thiazole-based peptides from marine resources: Structural and pharmacological aspects. Mar. Drugs 2020, 18, 329. [Google Scholar] [CrossRef]
- Garg, V.; Zhang, W.; Gidwani, P.; Kim, M.; Kolb, E.A. Preclinical analysis of tasidotin HCl in Ewing’s sarcoma, rhabdomyosarcoma, synovial sarcoma, and osteosarcoma. Clin. Cancer Res. 2007, 13, 5446–5454. [Google Scholar] [CrossRef]
- Bonate, P.L.; Beyerlein, D.; Crawford, J.; Roth, S.; Krumbholz, R.; Schmid, S. Pharmacokinetics in mice implanted with xenografted tumors after intravenous administration of tasidotin (ILX651) or its carboxylate metabolite. AAPS J. 2007, 9, 378–387. [Google Scholar] [CrossRef] [Green Version]
- Kaneda, M.; Sueyoshi, K.; Teruya, T.; Ohno, H.; Fujii, N.; Oishi, S. Total synthesis of odoamide, a novel cyclic depsipeptide, from an Okinawan marine cyanobacterium. Org. Biomol. Chem. 2016, 14, 9093–9104. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, M.M.; Ong, N.; Suggs, L. A general solid phase method for the synthesis of depsipeptides. Org. Biomol. Chem. 2013, 11, 1167–1170. [Google Scholar] [CrossRef] [Green Version]
- Campbell, T.D.; Febrian, R.; Kleinschmidt, H.E.; Smith, K.A.; Bracher, P.J. Quantitative Analysis of Glycine Oligomerization by Ion-Pair Chromatography. ACS Omega 2019, 4, 12745–12752. [Google Scholar] [CrossRef] [Green Version]
- Phyo, Y.; Ribeiro, J.; Fernandes, C.; Kijjoa, A.; Pinto, M.M.M. Marine natural peptides: Determination of absolute configuration using liquid chromatography methods and evaluation of bioactivities. Molecules 2018, 23, 306. [Google Scholar] [CrossRef] [Green Version]
- Greco, C.; Pfannenstiel, B.T.; Liu, J.C.; Keller, N.P. Depsipeptide Aspergillicins Revealed by Chromatin Reader Protein Deletion. ACS Chem. Biol. 2019, 14, 1121–1128. [Google Scholar] [CrossRef]
- Arumugam, V.; Venkatesan, M.; Ramachandran, S.; Sundaresan, U. Bioactive Peptides from Marine Ascidians and Future Drug Development–A Review. Int. J. Pept. Res. Ther. 2018, 24, 13–18. [Google Scholar] [CrossRef]
- Matsuo, Y.; Kanoh, K.; Imagawa, H.; Adachi, K.; Nishizawa, M.; Shizuri, Y. Urukthapelstatin A, a novel cytotoxic substance from marine-derived Mechercharimyces asporophorigenens YM11-542: II. Physico-chemical properties and structural elucidation. J. Antibiot. 2007, 60, 256–260. [Google Scholar] [CrossRef] [PubMed]
- Phyo, M.Y.; Katermeran, N.P.; Goh, J.X.; Tan, L.T. Trikoveramides A-C, cyclic depsipeptides from the marine cyanobacterium Symploca hydnoides. Phytochemistry 2021, 190, 112879. [Google Scholar] [CrossRef] [PubMed]
- Sable, G.A.; Park, J.; Kim, H.; Lim, S.J.; Jang, S.; Lim, D. Solid-Phase Total Synthesis of the Proposed Structure of Coibamide A and Its Derivative: Highly Methylated Cyclic Depsipeptides. Eur. J. Org. Chem. 2015, 2015, 7043–7052. [Google Scholar] [CrossRef]
- Kaur, H.; Harris, P.W.R.; Little, P.J.; Brimble, M.A. Total synthesis of the cyclic depsipeptide YM-280193, a platelet aggregation inhibitor. Org. Lett. 2015, 17, 492–495. [Google Scholar] [CrossRef] [PubMed]
- Pieszka, M.; Sobota, A.M.; Gačanin, J.; Weil, T.; Ng, D.Y.W. Orthogonally Stimulated Assembly/Disassembly of Depsipeptides by Rational Chemical Design. ChemBioChem 2019, 20, 1376–1381. [Google Scholar] [CrossRef]
- Lobo-Ruiz, A.; Tulla-Puche, J. General Fmoc-Based Solid-Phase Synthesis of Complex Depsipeptides Circumventing Problematic Fmoc Removal. Eur. J. Org. Chem. 2020, 2020, 183–192. [Google Scholar] [CrossRef]
- Jin, K.; Sam, I.H.; Po, K.H.L.; Lin, D.; Ghazvini Zadeh, E.H.; Chen, S.; Yuan, Y.; Li, X. Total synthesis of teixobactin. Nat. Commun. 2016, 7, 12394. [Google Scholar] [CrossRef] [Green Version]
- Merlino, F.; Tomassi, S.; Yousif, A.M.; Messere, A.; Marinelli, L.; Grieco, P.; Novellino, E.; Cosconati, S.; Di Maro, S. Boosting Fmoc Solid-Phase Peptide Synthesis by Ultrasonication. Org. Lett. 2019, 21, 6378–6382. [Google Scholar] [CrossRef]
- Farhid, H.; Rostami, M.M.; Shaabani, A.; Notash, B. Synthesis of Depsipeptides via Isocyanide-Based Consecutive Bargellini-Passerini Multicomponent Reactions. SynOpen 2021, 5, 167–172. [Google Scholar] [CrossRef]
- Batiste, S.M.; Johnston, J.N. Evidence for Ion-Templation during Macrocyclooligomerization of Depsipeptides. J. Am. Chem. Soc. 2018, 140, 4560–4568. [Google Scholar] [CrossRef]
- Jing, X.; Jin, K. A gold mine for drug discovery: Strategies to develop cyclic peptides into therapies. Med. Res. Rev. 2020, 40, 753–810. [Google Scholar] [CrossRef]
- Conte, M.; Fontana, E.; Nebbioso, A.; Altucci, L. Marine-derived secondary metabolites as promising epigenetic bio-compounds for anticancer therapy. Mar. Drugs 2021, 19, 15. [Google Scholar] [CrossRef]
- Eckes, K.M.; Baek, K.; Suggs, L.J. Design and Evaluation of Short Self-Assembling Depsipeptides as Bioactive and Biodegradable Hydrogels. ACS Omega 2018, 3, 1635–1644. [Google Scholar] [CrossRef] [Green Version]
- Ebenhan, T.; Mokaleng, B.B.; Venter, J.D.; Kruger, H.G.; Zeevaart, J.R.; Sathekge, M. Preclinical Assessment of a 68Ga-DOTA-Functionalized Depsipeptide as a Radiodiagnostic Infection Imaging Agent. Molecules 2017, 22, 1403. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Gong, X.; Li, P.; Lai, D.; Zhou, L. Structural diversity and biological activities of cyclic depsipeptides from fungi. Molecules 2018, 23, 169. [Google Scholar] [CrossRef] [Green Version]
- Bucci, R.; Dapiaggi, F.; Macut, H.; Pieraccini, S.; Sironi, M.; Gelmi, M.L.; Erba, E.; Pellegrino, S. On-resin multicomponent 1,3-dipolar cycloaddition of cyclopentanone–proline enamines and sulfonylazides as an efficient tool for the synthesis of amidino depsipeptide mimics. Amino Acids 2020, 52, 15–24. [Google Scholar] [CrossRef]
- Trinidad-Calderón, P.A.; Acosta-Cruz, E.; Rivero-Masante, M.N.; Díaz-Gómez, J.L.; García-Lara, S.; López-Castillo, L.M. Maize bioactive peptides: From structure to human health. J. Cereal Sci. 2021, 100, 103232. [Google Scholar] [CrossRef]
- De Vries, R.P.; Riley, R.; Wiebenga, A.; Aguilar-Osorio, G.; Amillis, S.; Uchima, C.A.; Anderluh, G.; Asadollahi, M.; Askin, M.; Barry, K.; et al. Comparative genomics reveals high biological diversity and specific adaptations in the industrially and medically important fungal genus Aspergillus. Genome Biol. 2017, 18, 28. [Google Scholar] [CrossRef] [Green Version]
- Keller, N.P. Translating biosynthetic gene clusters into fungal armor and weaponry. Nat. Chem. Biol. 2015, 11, 671–677. [Google Scholar] [CrossRef] [Green Version]
- Trinidad-Calderón, P.A.; López-Castillo, L.M.; Díaz-Gómez, J.L.; Soto, R.B.M.; Castorena-Torres, F.; García-Lara, S. Acetone-precipitated zein protein hydrolysates from blue-maize selectively target hepatocellular carcinoma and fibroblasts in a dose-dependent manner. Food Hydrocoll. Health 2022, 3, 100106. [Google Scholar] [CrossRef]
- Trinidad-Calderón, P.A.; Varela-Chinchilla, C.D.; García-Lara, S. Natural Peptides Inducing Cancer Cell Death: Mechanisms and Properties of Specific Candidates for Cancer Therapeutics. Molecules 2021, 26, 7453. [Google Scholar] [CrossRef] [PubMed]
- Lerman, I.; Hammes, S.R. Neutrophil elastase in the tumor microenvironment. Steroids 2018, 133, 96–101. [Google Scholar] [CrossRef] [PubMed]
- Dyshlovoy, S.A. Blue-print autophagy in 2020: A critical review. Mar. Drugs 2020, 18, 482. [Google Scholar] [CrossRef] [PubMed]
- Sone, H.; Shibata, T.; Fujita, T.; Ojika, M.; Yamada, K. Dolastatin H and isodolastatin H, potent cytotoxic peptides from the sea hare Dolabella auricularia: Isolation, stereostructures, and synthesis. J. Am. Chem. Soc. 1996, 118, 1874–1880. [Google Scholar] [CrossRef]
- Dougherty, P.G.; Sahni, A.; Pei, D. Understanding Cell Penetration of Cyclic Peptides. Chem. Rev. 2019, 119, 10241–10287. [Google Scholar] [CrossRef]
- Blanco-Míguez, A.; Gutiérrez-Jácome, A.; Pérez-Pérez, M.; Pérez-Rodríguez, G.; Catalán-García, S.; Fdez-Riverola, F.; Lourenço, A.; Sánchez, B. From amino acid sequence to bioactivity: The biomedical potential of antitumor peptides. Protein Sci. 2016, 25, 1084–1095. [Google Scholar] [CrossRef]
- Ghareeb, M.A.; Tammam, M.A.; El-Demerdash, A.; Atanasov, A.G. Insights about clinically approved and Preclinically investigated marine natural products. Curr. Res. Biotechnol. 2020, 2, 88–102. [Google Scholar] [CrossRef]
- Kulka, L.A.M.; Fangmann, P.V.; Panfilova, D.; Olzscha, H. Impact of HDAC Inhibitors on Protein Quality Control Systems: Consequences for Precision Medicine in Malignant Disease. Front. Cell Dev. Biol. 2020, 8, 425. [Google Scholar] [CrossRef]
- Aparicio Pelaz, D.; Yerkesh, Z.; Kirchgäßner, S.; Mahler, H.; Kharchenko, V.; Azhibek, D.; Jaremko, M.; Mootz, H.D.; Jaremko, Ł.; Schwarzer, D.; et al. Examining histone modification crosstalk using immobilized libraries established from ligation-ready nucleosomes. Chem. Sci. 2020, 11, 9218–9225. [Google Scholar] [CrossRef]
Key | Depsipeptide | IUPAC Condensed Formula | Molecular Weight (g/mol) | Ref. |
---|---|---|---|---|
a | Apratoxin | Cyclo[N(Me)Ala-N(Me)Ile-Pro-Unk-Tyr(Me)] | 840.1 | [31] |
b | Aurilide | Cyclo[N(Me)Ala-Unk-Val-D-N(Me)Leu-Sar-Val] | 834.1 | [21] |
c | Beauvericin | Cyclo[D-Oval-N(Me)Phe-D-Oval-N(Me)Phe-D-Oval-N(Me)Phe] | 783.9 | [32] |
d | Coibamide | N(Me2)Val-Oval-N(Me)Ser(Me)-N(Me)Leu-N(Me)Thr(1)-N(Me)Ser(Me)-N(Me)Ile-Ala-N(Me)Leu-Tyr(Me)-N(Me)Ala-(1) | 1287.6 | [33] |
e | Dehydrodidemnin | Pyruvoyl-Pro-D-N(Me)Leu-D-Thr(1)-Unk-Leu-Pro-DL-N(Me)Tyr(Me)-(1) | 1110.3 | [34] |
f | Enniatin | Cyclo[DL-Oval-DL-N(Me)xiIle-DL-Oval-DL-N(Me)xiIle-DL-Oval-DL-N(Me)xiIle] | 681.9 | [35] |
g | Grassypeptolide | Cyclo[D-N(Me)Leu-D-aThr-Unk-N(Me)Val-Pro-Unk] | 1102.4 | [36] |
h | Hantupeptin | Cyclo[N(Me)Ile-Ophe-Pro-N(Me)Val-Unk-Val] | 740.9 | [37] |
i | Lagunamide | Cyclo[Ala-D-N(Me)Phe-Sar-aIle-N(Me)Ala-Unk] | 842.1 | [38] |
j | Tiahuramide | Cyclo[N(Me)Ile-Unk-Val-N(Me)Val-Ophe-Pro] | 736.9 | [39] |
Depsipeptide | IUPAC Condensed Formula | Molecular Weight (g/mol) | Ref. |
---|---|---|---|
Beauvenniatin | Cyclo[D-OaIle-N(Me)Phe-D-OaIle-N(Me)Val-D-OaIle-N(Me)Val] | 729.9 | [54] |
Key | Depsipeptide | IUPAC Formula | Molecular Weight (g/mol) | Ref. |
---|---|---|---|---|
a | Micropeptins | (3S)-4-[[(2S,5S,8S,11R,12S,15S,18S,21R)-15-(4-aminobutyl)-2-[(2S)-butan-2-yl]-21-hydroxy-5-[(4-hydroxyphenyl)methyl]-4,11-dimethyl-3,6,9,13,16,22-hexaoxo-8-propan-2-yl-10-oxa-1,4,7,14,17-pentazabiCyclo [16.3.1]docosan-12-yl]amino]-3-(hexanoylamino)-4-oxobutanoic acid | 945.1 | [99] |
b | Tutuilamides | (2S)-N-[(2S,5S,8S,11R,12S,15Z,18S,21R)-2-benzyl-15-ethylidene-21-hydroxy-5-[(4-hydroxyphenyl)methyl]-4,11-dimethyl-3,6,9,13,16,22-hexaoxo-8-propan-2-yl-10-oxa-1,4,7,14,17-pentazabiCyclo [16.3.1]docosan-12-yl]-2-[[(2S)-2-[[I-4-chloro-3-methylbut-3-enoyl]amino]propanoyl]amino]-3-methylbutanamide | 1007.6 | [100] |
Key | Depsipeptide | IUPAC Formula | Molecular Weight (g/mol) | Ref. |
---|---|---|---|---|
a | Bassianolide | Cyclo[N(Me)Leu-D-Oval-N(Me)Leu-D-Oval-N(Me)Leu-D-Oval-N(Me)Leu-D-Oval] | 909.2 | [108] |
b | Clavatustide | Cyclo [2Abz-2Abz-D-Ophe-N(Et)Gly] | 471.5 | [109] |
c | Cryptophycin | 10-[(3-chloro-4-methoxyphenyl)methyl]-6-methyl-3-(2-methylpropyl)-16-[1-(3-phenyloxiran-2-yl)ethyl]-1,4-dioxa-8,11-diazacyclohexadec-13-ene-2,5,9,12-tetrone | 655.2 | [110] |
d | Largazole | S-[I-4-[(5R,8S,11S)-5-Me-6,9,13-trioxo-8-propan-2-yl-10-oxa-3,17-dithia-7,14,19,20-tetrazatriCyclo [14.2.1.12,5]icosa-1(18),2(20),16(19)-trien-11-yl]but-3-enyl] octanethioate | 622.9 | [111] |
e | Lyngbyabellin | (7S,14S,18S)-7-[(2S)-butan-2-yl]-14-(4,4-dichloropentyl)-18-(2-hydroxypropan-2-yl)-15,15-dimethyl-13,17-dioxa-9,20-dithia-3,6,22,23-tetrazatriCyclo [17.2.1.18,11]tricosa-1(21),8(23),10,19(22)-tetraene-2,5,12,16-tetrone | 691.7 | [18] |
f | Romidepsin | 7-ethylidene-4,21-di(propan-2-yl)-2-oxa-12,13-dithia-5,8,20,23-tetrazabiCyclo [8.7.6]tricos-16-ene-3,6,9,19,22-pentone | 540.7 | [112] |
g | Sansalvamide | Cyclo[L-leucyl-N-oxa-L-leucyl-L-valyl-L-leucyl-L-phenylalanyl] | 586.8 | [113] |
Key | Depsipeptide | IUPAC Formula | Molecular Weight (g/mol) | Ref. |
---|---|---|---|---|
a | Desmethoxymajusculamide C | Cyclo[Ala-Unk-Ala-Unk-Gly-N(Me)Ile-Gly-N(Me)Val-N(Me)Phe] | 955.2 | [142] |
b | Dolastatin | N(Me2)Val-Val-Unk | 785.1 | [143] |
c | Miuraenamide | (3E,15E)-6-[(3-bromo-4-hydroxyphenyl)methyl]-3-[methoxy(phenyl)methylidene]-7,9,16,19-tetramethyl-1-oxa-4,7,10-triazacyclononadec-15-ene-2,5,8,11-tetrone | 684.6 | [144] |
d | Nobilamide | Propionyl-D-Phe-D-Leu-Phe-D-aThr-Val-Ala-Abu(2,3-dehydro)-OH | 836.0 | [145] |
Key | Depsipeptide | IUPAC Formula | Molecular Weight (g/mol) | Ref. |
---|---|---|---|---|
a | Skyllamycins | N-[I-3-[2-[(Z)-1-Propenyl]phenyl]propenoyl]-Cyclo[L-Thr*-L-Ala-[(3S)-3-Me-L-Asp-]-Gly-[(βS)-β-hydroxy-L-Phe-]-L-Pro-[(βS)-β-hydroxy-O-Me-L-Tyr-]-D-Trp-[(2S)-2-hydroxy-Gly-]-D-Leu-[(3S)-3-hydroxy-D-Leu-]-] | 1483.6 | [160] |
b | Stereocalpin | Cyclo[Phe-N(Me)Phe-Unk] | 492.6 | [161] |
Key | Depsipeptide | IUPAC Formula | Molecular Weight (g/mol) | Ref. |
---|---|---|---|---|
a | Fusaristatin A | 3-[6,13-dimethyl-10-methylidene-2,5,9,12-tetraoxo-14-[(5E,7E)-3,7,11-trimethyl-4-oxoheptadeca-5,7-dienyl]-1-oxa-4,8,11-triazacyclotetradec-3-yl]propanamide | 658.9 | [170] |
b | N-methylsansalvamide | Cyclo[Leu-Oleu-Val-N(Me)Leu-Phe] | 600.8 | [28] |
Depsipeptide | IUPAC Formula | Molecular Weight (g/mol) | Ref. |
---|---|---|---|
Kahalalide F | L-Val, N-(5-Me-1-oxohexyl)-D-valyl-l-threonyl-l-valyl-D-valyl-D-prolyl-l-ornithyl-D-alloisoleucyl-D-allothreonyl-D-alloisoleucyl-D-valyl-l-phenylalanyl-(2z)-2-amino-2-butenoyl-, (13->8)-lactone | 1477.9 | [180] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Trinidad-Calderón, P.A.; Varela-Chinchilla, C.D.; García-Lara, S. Depsipeptides Targeting Tumor Cells: Milestones from In Vitro to Clinical Trials. Molecules 2023, 28, 670. https://doi.org/10.3390/molecules28020670
Trinidad-Calderón PA, Varela-Chinchilla CD, García-Lara S. Depsipeptides Targeting Tumor Cells: Milestones from In Vitro to Clinical Trials. Molecules. 2023; 28(2):670. https://doi.org/10.3390/molecules28020670
Chicago/Turabian StyleTrinidad-Calderón, Plinio A., Carlos Daniel Varela-Chinchilla, and Silverio García-Lara. 2023. "Depsipeptides Targeting Tumor Cells: Milestones from In Vitro to Clinical Trials" Molecules 28, no. 2: 670. https://doi.org/10.3390/molecules28020670
APA StyleTrinidad-Calderón, P. A., Varela-Chinchilla, C. D., & García-Lara, S. (2023). Depsipeptides Targeting Tumor Cells: Milestones from In Vitro to Clinical Trials. Molecules, 28(2), 670. https://doi.org/10.3390/molecules28020670