1
|
Xi M, Zhu J, Zhang F, Shen H, Chen J, Xiao Z, Huangfu Y, Wu C, Sun H, Xia G. Antibody-drug conjugates for targeted cancer therapy: Recent advances in potential payloads. Eur J Med Chem 2024; 276:116709. [PMID: 39068862 DOI: 10.1016/j.ejmech.2024.116709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a promising cancer therapy modality which specifically delivers highly toxic payloads to cancer cells through antigen-specific monoclonal antibodies (mAbs). To date, 15 ADCs have been approved and more than 100 ADC candidates have advanced to clinical trials for the treatment of various cancers. Among these ADCs, microtubule-targeting and DNA-damaging agents are at the forefront of payload development. However, several challenges including toxicity and drug resistance limit the potential of this modality. To tackle these issues, multiple innovative payloads such as immunomodulators and proteolysis targeting chimeras (PROTACs) are incorporated into ADCs to enable multimodal cancer therapy. In this review, we describe the mechanism of ADCs, highlight the importance of ADC payloads and summarize recent progresses of conventional and unconventional ADC payloads, trying to provide an insight into payload diversification as a key step in future ADC development.
Collapse
Affiliation(s)
- Meiyang Xi
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing, 312000, China; College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Jingjing Zhu
- NovoCodex Biopharmaceuticals Co. Ltd., Shaoxing, 312090, China
| | - Fengxia Zhang
- NovoCodex Biopharmaceuticals Co. Ltd., Shaoxing, 312090, China
| | - Hualiang Shen
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing, 312000, China; College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Jianhui Chen
- College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Ziyan Xiao
- College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Yanping Huangfu
- College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Chunlei Wu
- Zhejiang Engineering Research Center of Fat-soluble Vitamin, Shaoxing University, Shaoxing, 312000, China; College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing, 312000, China
| | - Haopeng Sun
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China.
| | - Gang Xia
- NovoCodex Biopharmaceuticals Co. Ltd., Shaoxing, 312090, China
| |
Collapse
|
2
|
Wang Y, Song Y, He Y, Wang Y, Maurer J, Kiessling F, Lammers T, Wang F, Shi Y. Direct immunoactivation by chemotherapeutic drugs in cancer treatment. ADVANCED THERAPEUTICS 2023; 6:2300209. [PMID: 38249990 PMCID: PMC7615547 DOI: 10.1002/adtp.202300209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Indexed: 01/23/2024]
Abstract
The immune system plays a crucial role in recognizing and eliminating pathogenic substances and malignant cells in the body. For cancer treatment, immunotherapy is becoming the standard treatment for many types of cancer and is often combined with chemotherapy. Although chemotherapeutic agents are often reported to have adverse effects, including immunosuppression, they can also play a positive role in immunotherapy by directly stimulating the immune system. This has been demonstrated in preclinical and clinical studies in the past decades. Chemotherapeutics can activate immune cells through different immune receptors and signaling pathways depending on their chemical structure and formulation. In this review, we summarize and discuss the direct immunoactivation effects of chemotherapeutics and possible mechanisms behind these effects. Finally, we prospect chemo-immunotherapeutic combinations for the more effective and safer treatment of cancer.
Collapse
Affiliation(s)
- Yurui Wang
- Department of Polymer Therapeutics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Yiran Song
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Yazhi He
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Yang Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Jochen Maurer
- Department of Gynecology and Obstetrics, Uniklinik RWTH Aachen, Aachen 52074, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Feng Wang
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, PR China
| | - Yang Shi
- Department of Polymer Therapeutics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| |
Collapse
|
3
|
Wang Z, Li H, Gou L, Li W, Wang Y. Antibody-drug conjugates: Recent advances in payloads. Acta Pharm Sin B 2023; 13:4025-4059. [PMID: 37799390 PMCID: PMC10547921 DOI: 10.1016/j.apsb.2023.06.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/30/2023] [Accepted: 06/23/2023] [Indexed: 10/05/2023] Open
Abstract
Antibody‒drug conjugates (ADCs), which combine the advantages of monoclonal antibodies with precise targeting and payloads with efficient killing, show great clinical therapeutic value. The ADCs' payloads play a key role in determining the efficacy of ADC drugs and thus have attracted great attention in the field. An ideal ADC payload should possess sufficient toxicity, low immunogenicity, high stability, and modifiable functional groups. Common ADC payloads include tubulin inhibitors and DNA damaging agents, with tubulin inhibitors accounting for more than half of the ADC drugs in clinical development. However, due to clinical limitations of traditional ADC payloads, such as inadequate efficacy and the development of acquired drug resistance, novel highly efficient payloads with diverse targets and reduced side effects are being developed. This perspective summarizes the recent research advances of traditional and novel ADC payloads with main focuses on the structure-activity relationship studies, co-crystal structures, and designing strategies, and further discusses the future research directions of ADC payloads. This review also aims to provide valuable references and future directions for the development of novel ADC payloads that will have high efficacy, low toxicity, adequate stability, and abilities to overcome drug resistance.
Collapse
Affiliation(s)
- Zhijia Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| | - Hanxuan Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Lantu Gou
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| |
Collapse
|
4
|
Singh SB. Discovery and Development of Dolastatin 10-Derived Antibody Drug Conjugate Anticancer Drugs. JOURNAL OF NATURAL PRODUCTS 2022; 85:666-687. [PMID: 35072477 DOI: 10.1021/acs.jnatprod.1c01135] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Dolastatin 10 is an extremely potent broad-spectrum antitubulin anticancer pentapeptide isolated from Dolabella auricularia. The two-dimensional structure was elucidated by NMR and mass spectrometric analyses. The absolute configuration was determined by a convergent total synthesis. SAR studies established that modifications at C- and N-terminals were tolerated for cytotoxic activity. Human clinical trials of dolastatin 10 and auristatin PE (a C-terminal analog) showed occasional signs of efficacy but failed due to lack of separation of toxicity and efficacy. Nanomolar cytotoxicity helped transition this class of pentapeptides to the next phase of development as antibody drug conjugates (ADCs) by reducing systemic toxicity. Four ADC drugs (Adcetris, Padcev, Polivy, and Blenrep) carrying monomethyl auristatin E (MMAE, vedotin) and monomethyl auristatin F (MMAF, mafodotin) payloads have been approved for treatment of a number of cancers expressing antibody-specific antigens. More than 36 ADCs carrying a variety of pentapeptide analogues are undergoing preclinical and clinical developments. They are being evaluated in more than 200 human trials. A comprehensive review of the discovery, total synthesis of dolastatin 10 and new amino acids, SAR studies of dolastatin 10 and auristatins, conjugations to antibodies, and preclinical and clinical development of ADCs have been presented.
Collapse
Affiliation(s)
- Sheo B Singh
- SBS Pharma Consulting LLC, Edison, New Jersey 08820, United States
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| |
Collapse
|
5
|
Mbaoji FN, Nweze JA, Yang L, Huang Y, Huang S, Onwuka AM, Peter IE, Mbaoji CC, Jiang M, Zhang Y, Pan L, Yang D. Novel Marine Secondary Metabolites Worthy of Development as Anticancer Agents: A Review. Molecules 2021; 26:molecules26195769. [PMID: 34641312 PMCID: PMC8510081 DOI: 10.3390/molecules26195769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022] Open
Abstract
Secondary metabolites from marine sources have a wide range of biological activity. Marine natural products are promising candidates for lead pharmacological compounds to treat diseases that plague humans, including cancer. Cancer is a life-threatening disorder that has been difficult to overcome. It is a long-term illness that affects both young and old people. In recent years, significant attempts have been made to identify new anticancer drugs, as the existing drugs have been useless due to resistance of the malignant cells. Natural products derived from marine sources have been tested for their anticancer activity using a variety of cancer cell lines derived from humans and other sources, some of which have already been approved for clinical use, while some others are still being tested. These compounds can assault cancer cells via a variety of mechanisms, but certain cancer cells are resistant to them. As a result, the goal of this review was to look into the anticancer potential of marine natural products or their derivatives that were isolated from January 2019 to March 2020, in cancer cell lines, with a focus on the class and type of isolated compounds, source and location of isolation, cancer cell line type, and potency (IC50 values) of the isolated compounds that could be a guide for drug development.
Collapse
Affiliation(s)
- Florence Nwakaego Mbaoji
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning 530007, China; (F.N.M.); (J.A.N.); (Y.H.); (S.H.)
- College of Life Science and Technology of Guangxi University, Nanning 530004, China
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Enugu State, Nigeria; (A.M.O.); (I.E.P.); (C.C.M.)
| | - Justus Amuche Nweze
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning 530007, China; (F.N.M.); (J.A.N.); (Y.H.); (S.H.)
- Department of Science Laboratory Technology, Faculty of Physical Sciences, University of Nigeria, Nsukka 410001, Enugu State, Nigeria
- Department of Ecosystem Biology, Faculty of Science, University of South Bohemia in Ceske Budejovice, 37005 Ceske Budejovice, Czech Republic
- Soil and Water Research Infrastructure, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| | - Liyan Yang
- Guangxi Biomass Industrialization Engineering Institute, National Engineering Research Center of Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass, Guangxi Academy of Sciences, Nanning 530007, China;
| | - Yangbin Huang
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning 530007, China; (F.N.M.); (J.A.N.); (Y.H.); (S.H.)
| | - Shushi Huang
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning 530007, China; (F.N.M.); (J.A.N.); (Y.H.); (S.H.)
| | - Akachukwu Marytheresa Onwuka
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Enugu State, Nigeria; (A.M.O.); (I.E.P.); (C.C.M.)
| | - Ikechukwu Emmanuel Peter
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Enugu State, Nigeria; (A.M.O.); (I.E.P.); (C.C.M.)
| | - Cynthia Chioma Mbaoji
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Enugu State, Nigeria; (A.M.O.); (I.E.P.); (C.C.M.)
| | - Mingguo Jiang
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi University for Nationalities, Nanning 530008, China;
| | - Yunkai Zhang
- College of Life Science and Technology of Guangxi University, Nanning 530004, China
- Correspondence: (Y.Z.); (L.P.); (D.Y.); Tel.: +86-771-2503980 (L.P.); +86-771-2536109 (D.Y.)
| | - Lixia Pan
- Guangxi Biomass Industrialization Engineering Institute, National Engineering Research Center of Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass, Guangxi Academy of Sciences, Nanning 530007, China;
- Correspondence: (Y.Z.); (L.P.); (D.Y.); Tel.: +86-771-2503980 (L.P.); +86-771-2536109 (D.Y.)
| | - Dengfeng Yang
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning 530007, China; (F.N.M.); (J.A.N.); (Y.H.); (S.H.)
- Correspondence: (Y.Z.); (L.P.); (D.Y.); Tel.: +86-771-2503980 (L.P.); +86-771-2536109 (D.Y.)
| |
Collapse
|
6
|
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. [PMID: 34202685 PMCID: PMC8303260 DOI: 10.3390/md19070363] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/19/2021] [Accepted: 06/20/2021] [Indexed: 12/22/2022] Open
Abstract
Dolastatin 10 (Dol-10), a leading marine pentapeptide isolated from the Indian Ocean mollusk Dolabella auricularia, contains three unique amino acid residues. Dol-10 can effectively induce apoptosis of lung cancer cells and other tumor cells at nanomolar concentration, and it has been developed into commercial drugs for treating some specific lymphomas, so it has received wide attention in recent years. In vitro experiments showed that Dol-10 and its derivatives were highly lethal to common tumor cells, such as L1210 leukemia cells (IC50 = 0.03 nM), small cell lung cancer NCI-H69 cells (IC50 = 0.059 nM), and human prostate cancer DU-145 cells (IC50 = 0.5 nM), etc. With the rise of antibody-drug conjugates (ADCs), milestone progress was made in clinical research based on Dol-10. A variety of ADCs constructed by combining MMAE or MMAF (Dol-10 derivatives) with a specific antibody not only ensured the antitumor activity of the drugs themself but also improved their tumor targeting and reduced the systemic toxicity. They are currently undergoing clinical trials or have been approved for marketing, such as Adcetris®, which had been approved for the treatment of anaplastic large T-cell systemic malignant lymphoma and Hodgkin lymphoma. Dol-10, as one of the most medically valuable natural compounds discovered up to now, has brought unprecedented hope for tumor treatment. It is particularly noteworthy that, by modifying the chemical structure of Dol-10 and combining with the application of ADCs technology, Dol-10 as a new drug candidate still has great potential for development. In this review, the biological activity and chemical work of Dol-10 in the advance of antitumor drugs in the last 35 years will be summarized, which will provide the support for pharmaceutical researchers interested in leading exploration of antitumor marine peptides.
Collapse
Affiliation(s)
- Gang Gao
- School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo 315211, China;
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China; (H.H.); (D.L.)
| | - Yanbing Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China;
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China; (H.H.); (D.L.)
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China; (H.H.); (D.L.)
| | - Chunlan Tang
- School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo 315211, China;
| |
Collapse
|
7
|
Newman DJ. Natural Product Based Antibody Drug Conjugates: Clinical Status as of November 9, 2020. JOURNAL OF NATURAL PRODUCTS 2021; 84:917-931. [PMID: 33635651 DOI: 10.1021/acs.jnatprod.1c00065] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
As of early November 2020, there are 10 approved antibody drug conjugates (ADCs) plus two others that are not usually listed. In addition, there are 70 ADCs at stages from phase I to phase III and 23 that are at the preclinical stage. The warheads of all of these drugs and drug candidates have their origins in natural product structures. The sources and modifications are discussed in general and then specifically commented on in each case with either the generic name if known and/or the ADC's ID names. Interestingly, almost all warheads listed are from microbial sources though initially a number were thought to have been from plants. The latest NCT numbers from Clintrials.gov of all phase I to phase III candidates are also given. Three unusual ADCs are also discussed, two of which (an antitumor agent and one directed against autoimmune diseases) are not usually listed as ADCs, with the third being an anti-infective.
Collapse
Affiliation(s)
- David J Newman
- NIH Special Volunteer, Wayne, Pennsylvania 19087, United States
| |
Collapse
|
8
|
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:E247. [PMID: 33466486 PMCID: PMC7796498 DOI: 10.3390/molecules26010247] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022] Open
Abstract
Cyanobacteria (blue-green microalgae) are ubiquitous, Gram-negative photoautotrophic prokaryotes. They are considered as one of the most efficient sources of bioactive secondary metabolites. More than 50% of cyanobacteria are cultivated on commercial platforms to extract bioactive compounds, which have bene shown to possess anticancer activity. The chemically diverse natural compounds or their analogues induce cytotoxicity and potentially kill a variety of cancer cells via the induction of apoptosis, or altering the activation of cell signaling, involving especially the protein kinase-C family members, cell cycle arrest, mitochondrial dysfunctions and oxidative damage. These therapeutic properties enable their use in the pharma and healthcare sectors for the betterment of future generations. This review provides a baseline overview of the anti-cancerous cyanobacterial bioactive compounds, along with recently introduced nanomaterials that could be used for the development of new anticancer drugs to build a healthy future for mankind.
Collapse
Affiliation(s)
- Hina Qamar
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India;
| | - Kashif Hussain
- Pharmacy Section, Gyani Inder Singh Institute of Professional Studies, Dehradun 248003, India;
- School of Pharmacy, Glocal University, Saharanpur 247121, India
| | - Aishwarya Soni
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal, Sonepat 124001, India;
| | - Anish Khan
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak 124001, India;
| | - Touseef Hussain
- Department of Botany, Aligarh Muslim University, Aligarh 202002, India
| | - Benoît Chénais
- EA 2160 Mer Molécules Santé, Le Mans Université, F-72085 Le Mans, France
| |
Collapse
|
9
|
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. [PMID: 32652435 DOI: 10.1016/j.ejmech.2020.112473] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/27/2022]
Abstract
Cyanobacteria are one of the oldest creatures on earth, originated 3.5-3.3 billion years ago, and are distributed all over the world, including freshwater ponds and lakes, hot springs, and polar ice, especially in tropical and subtropical marine locations. Due to their large multimodular non-ribosomal peptide synthetases (NRPS) and polyketide synthases (PKS) biosynthetic machinery, cyanobacteria have represented a significant new source of structurally bioactive secondary metabolites. Moorea as a prolific producer have yielded lots of natural products with a variety of bioactivities such as highly cytotoxicity, anticancer activity, ion channel blocking activity, brine shrimp toxicity and other activities. Some of secondary metabolites have been identified as potential lead compounds for the development of anticancer agents. In this review, a total of 111 bioactive marine cyanobacterial secondary metabolites from the genus Moorea, published in the 54 literatures updated to the middle of 2019 and some synthetic analogues, are discussed with emphasis on their structures and biological activities.
Collapse
Affiliation(s)
- Jianzhou Xu
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Department of Marine Pharmacy, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315832, China
| | - Ting Zhang
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Department of Marine Pharmacy, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315832, China
| | - Jiaxiao Yao
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Department of Marine Pharmacy, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315832, China
| | - Jian Lu
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Department of Marine Pharmacy, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315832, China
| | - Zhiwen Liu
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Department of Marine Pharmacy, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315832, China
| | - Lijian Ding
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Department of Marine Pharmacy, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315832, China.
| |
Collapse
|
10
|
Pettit GR, Melody N, Chapuis JC. Antineoplastic Agents. 607. Emetine Auristatins. JOURNAL OF NATURAL PRODUCTS 2020; 83:1571-1576. [PMID: 32323990 DOI: 10.1021/acs.jnatprod.0c00031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The remarkable biological activity of the dolastatin 10 structural modifications quinstatins and isoquinstatins prompted further investigation into drug hybrids containing biologically active isoquinoline moieties. In this study, the isoquinoline alkaloid emetine was selected as one of the structural domains of a hybrid molecule. That was accomplished by covalently bonding the Dov-Val-Dil-Dap peptide sequence of dolastatin 10 peptide at the N-2' secondary amine of emetine. Three new hybrids were synthesized, 5, 9, and 10. Comparison of the biological activity of these new peptide-emetine analogues with emetine showed complete retention of activity for 5 and a 10-fold decrease for hybrids 9 and 10. The result was surprising, as the activity of emetine is usually lost or greatly reduced when substituted at the N-2' position.
Collapse
Affiliation(s)
- George R Pettit
- School of Molecular Sciences, Arizona State University, P.O. Box 871604, Tempe, Arizona 85287-1604, United States
| | - Noeleen Melody
- School of Molecular Sciences, Arizona State University, P.O. Box 871604, Tempe, Arizona 85287-1604, United States
| | - Jean-Charles Chapuis
- School of Molecular Sciences, Arizona State University, P.O. Box 871604, Tempe, Arizona 85287-1604, United States
| |
Collapse
|
11
|
Aaghaz S, Gohel V, Kamal A. Peptides as Potential Anticancer Agents. Curr Top Med Chem 2019; 19:1491-1511. [DOI: 10.2174/1568026619666190125161517] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 12/26/2018] [Accepted: 01/18/2019] [Indexed: 12/21/2022]
Abstract
Cancer consists of heterogeneous multiple cell subpopulation which at a later stage develop resistant phenotypes, which include resistance to pro-apoptotic stimuli and/or cytotoxic resistance to anticancer compounds. The property of cancerous cells to affect almost any part of the body categorizes cancer to many anatomic and molecular subtypes, each requiring a particular therapeutic intervention. As several modalities are hindered in a variety of cancers and as the cancer cells accrue varied types of oncogenic mutations during their progression the most likely benefit will be obtained by a combination of therapeutic agents that might address the diverse hallmarks of cancer. Natural compounds are the backbone of cancer therapeutics owing to their property of affecting the DNA impairment and restoration mechanisms and also the gene expression modulated via several epigenetic molecular mechanisms. Bioactive peptides isolated from flora and fauna have transformed the arena of antitumour therapy and prompt progress in preclinical studies is promising. The difficulties in creating ACP rest in improving its delivery to the tumour site and it also must maintain a low toxicity profile. The substantial production costs, low selectivity and proteolytic stability of some ACP are some of the factors hindering the progress of peptide drug development. Recently, several publications have tried to edify the field with the idea of using peptides as adjuvants with established drugs for antineoplastic use. This review focuses on peptides from natural sources that precisely target tumour cells and subsequently serve as anticancer agents that are less toxic to normal tissues.
Collapse
Affiliation(s)
- Shams Aaghaz
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S Nagar, Mohali, India
| | - Vivek Gohel
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S Nagar, Mohali, India
| | - Ahmed Kamal
- School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, India
| |
Collapse
|
12
|
Guo Z, Shi H, Li C, Luo Y, Bi S, Yu R, Wang H, Liu W, Zhu J, Huang W, Song L. Identification and Characterization of a Novel Protein ASP-3 Purified from Arca subcrenata and Its Antitumor Mechanism. Mar Drugs 2019; 17:E528. [PMID: 31505835 PMCID: PMC6780846 DOI: 10.3390/md17090528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 01/04/2023] Open
Abstract
Diverse bioactive substances derived from marine organisms have been attracting growing attention. Besides small molecules and polypeptides, numerous studies have shown that marine proteins also exhibit antitumor activities. Small anticancer proteins can be expressed in vivo by viral vectors to exert local and long-term anticancer effects. Herein, we purified and characterized a novel protein (ASP-3) with unique antitumor activity from Arca subcrenata Lischke. The ASP-3 contains 179 amino acids with a molecular weight of 20.6 kDa. The spectral characterization of ASP-3 was elucidated using Fourier Transform infrared spectroscopy (FTIR) and Circular Dichroism (CD) spectroscopy. Being identified as a sarcoplasmic calcium-binding protein, ASP-3 exhibited strong inhibitory effects on the proliferation of Human hepatocellular carcinoma (HepG2) cells with an IC50 value of 171.18 ± 18.59 μg/mL, measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The RNA-seq analysis showed that ASP-3 regulated the vascular endothelial growth factor receptor (VEGFR) signaling pathway in HepG2 cells. Immunofluorescence results indicated that ASP-3 effectively reduced VEGFR2 phosphorylation in HepG2 cells and affected the downstream components of VEGF signaling pathways. The surface plasmon resonance (SPR) analysis further demonstrated that ASP-3 direct interacted with VEGFR2. More importantly, the therapeutic potential of ASP-3 as an anti-angiogenesis agent was further confirmed by an in vitro model using VEGF-induced tube formation assay of human umbilical vein endothelial cells (HUVECs), as well as an in vivo model using transgenic zebrafish model. Taken together, the ASP-3 provides a good framework for the development of even more potent anticancer proteins and provides important weapon for cancer treatment using novel approaches such as gene therapy.
Collapse
Affiliation(s)
- Zhongyi Guo
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou 510632, China.
- Center for experimental technology, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Hui Shi
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China.
| | - Chunlei Li
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China.
| | - Yuanyuan Luo
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China.
| | - Sixue Bi
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Rongmin Yu
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China.
| | - Haoran Wang
- Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA.
| | - Wanying Liu
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China.
| | - Jianhua Zhu
- Biotechnological Institute of Chinese Materia Medica, Jinan University, Guangzhou 510632, China.
| | - Weijuan Huang
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Liyan Song
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
13
|
Wali AF, Majid S, Rasool S, Shehada SB, Abdulkareem SK, Firdous A, Beigh S, Shakeel S, Mushtaq S, Akbar I, Madhkali H, Rehman MU. Natural products against cancer: Review on phytochemicals from marine sources in preventing cancer. Saudi Pharm J 2019; 27:767-777. [PMID: 31516319 PMCID: PMC6733955 DOI: 10.1016/j.jsps.2019.04.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/20/2019] [Indexed: 12/14/2022] Open
Abstract
Marine natural products have as of now been acknowledged as the most important source of bioactive substances and drug leads. Marine flora and fauna, such as algae, bacteria, sponges, fungi, seaweeds, corals, diatoms, ascidian etc. are important resources from oceans, accounting for more than 90% of the total oceanic biomass. They are taxonomically different with huge productive and are pharmacologically active novel chemical signatures and bid a tremendous opportunity for discovery of new anti-cancer molecules. The water bodies a rich source of potent molecules which improve existence suitability and serve as chemical shield against microbes and little or huge creatures. These molecules have exhibited a range of biological properties antioxidant, antibacterial, antitumour etc. In spite of huge resources enriched with exciting chemicals, the marine floras and faunas are largely unexplored for their anticancer properties. In recent past, numerous marine anticancer compounds have been isolated, characterized, identified and are under trials for human use. In this write up we have tried to compile about marine-derived compounds anticancer biological activities of diverse flora and fauna and their underlying mechanisms and the generous raise in these compounds examined for malignant growth treatment in the course of the most recent quite a long while.
Collapse
Affiliation(s)
- Adil Farooq Wali
- RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Sabhiya Majid
- Department of Biochemistry, Govt. Medical College (GMC), Karan Nagar, Srinagar 190010, J&K, India
| | - Shabhat Rasool
- Department of Biochemistry, Govt. Medical College (GMC), Karan Nagar, Srinagar 190010, J&K, India
| | - Samar Bassam Shehada
- RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Shahad Khalid Abdulkareem
- RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Aimen Firdous
- Department of Processing Technology, Kerala University of Fisheries and Ocean Studies (KUFOS), Panangad 682506, Kerala, India
| | - Saba Beigh
- Institut de Biologie, Molecular et Cellulaire, CNRS, immunopathologie et Chimie Therapeutique, Strasbourg Cedex, France
| | - Sheeba Shakeel
- Department of Pharmaceutical Sciences, Faculty of Applied Sciences, University of Kashmir, Srinagar 110006, J&K, India
| | - Saima Mushtaq
- Faculty of Veterinary Science and Animal Husbandry, SKUAST-Kashmir, Shuhama 190006, J&K, India
| | - Imra Akbar
- School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Hassan Madhkali
- Department of Pharmacology, College of Pharmacy, Prince Sattan Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Muneeb U. Rehman
- Department of Biochemistry, Govt. Medical College (GMC), Karan Nagar, Srinagar 190010, J&K, India
| |
Collapse
|
14
|
Yang K, Chen B, Gianolio DA, Stefano JE, Busch M, Manning C, Alving K, Gregory RC, Brondyk WH, Miller RJ, Dhal PK. Convergent synthesis of hydrophilic monomethyl dolastatin 10 based drug linkers for antibody-drug conjugation. Org Biomol Chem 2019; 17:8115-8124. [PMID: 31460552 DOI: 10.1039/c9ob01639b] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We report a modular approach to synthesize maleimido group containing hydrophilic dolastatin 10 (Dol10) derivatives as drug-linkers for the syntheses of antibody-drug conjugates (ADCs). Discrete polyethylene glycol (PEG) moieties of different chain lengths were introduced as part of the linker to impart hydrophilicity to these drug linkers. The synthesis process involved construction of PEG maleimido derivatives of the tetrapeptide intermediate (N-methylvaline-valine-dolaisoleucine-dolaproine), which were subsequently coupled with dolaphenine to generate the desired drug linkers. The synthetic method reported in this manuscript circumvents the use of highly cytotoxic Dol10 in its native form. By using trastuzumab (Herceptin®) as the antibody we have synthesized Dol10 containing ADCs. The presence of a discrete PEG chain in the drug linkers resulted in ADCs free from aggregation. The effect of PEG chain length on the biological activities of these Dol10 containing ADCs was investigated by in vitro cytotoxicity assays. ADCs containing PEG6 and PEG8 spacers exhibited the highest level of in vitro anti-proliferative activity against HER2-positive (SK-BR-3) human tumor cells. ADCs derived from Herceptin® and PEG8-Dol10, at a dose of 10 mg kg-1, effectively delayed the tumor growth and prolonged the survival time in mice bearing human ovarian SKOV-3 xenografts.
Collapse
Affiliation(s)
- Kanwen Yang
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | - Bo Chen
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | | | - James E Stefano
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | - Michelle Busch
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | | | - Kim Alving
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | | | | | - Robert J Miller
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | - Pradeep K Dhal
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| |
Collapse
|
15
|
The "Utility" of Highly Toxic Marine-Sourced Compounds. Mar Drugs 2019; 17:md17060324. [PMID: 31159276 PMCID: PMC6627392 DOI: 10.3390/md17060324] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 05/22/2019] [Accepted: 05/27/2019] [Indexed: 12/27/2022] Open
Abstract
Currently a few compounds isolated from marine sources have become drugs, mainly directed towards cancer and pain. Compounds from marine sources have exquisite potencies against eukaryotic cells, as they act as protective agents against attack by predators in the marine environment. Their toxicities act as a “double-edged sword” as they are often too toxic for direct use in humans and thus have to be chemically modified. By linking suitably modified compounds to monoclonal antibodies directed against specific epitopes in mammalian cancer cells, they can be delivered to a specific cell type in humans. This review updates and extends an article published in early 2017, demonstrating how by careful chemical modifications, highly toxic compounds, frequently peptidic in nature, can be utilized as antitumor drug candidates. The antibody-drug- conjugates (ADCs) discussed are those that are currently in clinical trials listed in the NIH Clinical Trials Registry as, “currently active, recruiting or in some cases, recently completed”. There are also some ADCs discussed that are at the advanced preclinical stage, that in some cases, are repurposing current drug entities, and the review finishes with a short discussion of the aplyronines as potential candidate warheads as a result of scalable synthetic processes.
Collapse
|
16
|
Xing H, Tong M, Jiang N, Zhang X, Hu H, Pan H, Li D. Antitumour bioactive peptides isolated from marine organisms. Clin Exp Pharmacol Physiol 2018; 44:1077-1082. [PMID: 28675498 DOI: 10.1111/1440-1681.12808] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 06/28/2017] [Accepted: 06/28/2017] [Indexed: 11/30/2022]
Abstract
Marine organisms are an important source of antitumour active substances. Thus, pharmaceutical research in recent years has focused on exploring new antitumour drugs derived from marine organisms, and, many peptide drugs with strong antitumour activities have been successfully extracted. Based on different mechanisms, this paper reviews the research on several typical antitumour bioactive peptides in marine drugs and the latest progress therein. Additionally, the development prospects for these antitumour bioactive peptide-based drugs are discussed so as to provide a reference for future research in this field.
Collapse
Affiliation(s)
- Haibo Xing
- Department of ICU, Xiasha Campus, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Mengting Tong
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Nanyu Jiang
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaomin Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Hong Hu
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Da Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Pettit GR, Melody N, Chapuis JC. Antineoplastic Agents. 605. Isoquinstatins. JOURNAL OF NATURAL PRODUCTS 2018; 81:451-457. [PMID: 28926240 DOI: 10.1021/acs.jnatprod.7b00352] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In order to further explore quinoline-type structural modification of the powerful anticancer drug dolastatin 10, an Indian Ocean sea hare constituent and parent molecule of the very successful antibody drug conjugate (ADC) Adcetris, our recent quinstatin study has been extended by replacing the quinoline ring with an isoquinoline. The resulting isoquinstatins (4-6) were modified to N-terminal desmethylisoquinstatins (7-9) and, in turn, bonded to appropriate linker units to give linker-desmethylisoquinstatin conjugates 11-13 in preparation for eventual monoclonal antibody attachment. Comparison of the new isoquinstatins with their quinstatin counterparts against six human cancer cell lines indicated the isoquinstatins to have GI50 values that were comparable to or somewhat higher than those of the isomeric quinstatins. However, desmethylisoquinstatin 5 (7) was significantly more potent than its desmethylquinstatin 5 analogue. When evaluated against quinstatin 8, its isoquinstatin 8 (6) counterpart was somewhat less potent. In general, the isoquinstatins evaluated proved to be quite strong cancer cell growth inhibitors.
Collapse
Affiliation(s)
- George R Pettit
- Department of Chemistry and Biochemistry , Arizona State University , P.O. Box 871604, Tempe , Arizona 85287-1604 , United States
| | - Noeleen Melody
- Department of Chemistry and Biochemistry , Arizona State University , P.O. Box 871604, Tempe , Arizona 85287-1604 , United States
| | - Jean-Charles Chapuis
- Department of Chemistry and Biochemistry , Arizona State University , P.O. Box 871604, Tempe , Arizona 85287-1604 , United States
| |
Collapse
|
18
|
Pettit GR, Melody N, Chapuis JC. Antineoplastic Agents. 604. The Path of Quinstatin Derivatives to Antibody Drug Conjugates. JOURNAL OF NATURAL PRODUCTS 2017; 80:2447-2452. [PMID: 28895394 DOI: 10.1021/acs.jnatprod.7b00237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
To further evaluate the exceptional cancer cell growth inhibition by the quinstatins, of which one of the series, quinstatin 8, approaches the exceptional cytotoxic activity of the parent dolastatin 10 (1), four of the quinstatins have been converted to desmethyl derivatives. Three of the four (4, 5, and 8 [7b-d]) were next bonded to the linker (8) employed in the synthesis of the very successful and structurally related anticancer drug Adcetris (3). Owing to these structural modifications, a next step could be taken by bonding to a monoclonal antibody, thereby producing an antibody drug conjugate (ADC) related to Adcetris structurally but with the possibility of a wider spectrum of activity and utility.
Collapse
Affiliation(s)
- George R Pettit
- Department of Chemistry and Biochemistry, Arizona State University , P.O. Box 871604, Tempe, Arizona 85287-1604, United States
| | - Noeleen Melody
- Department of Chemistry and Biochemistry, Arizona State University , P.O. Box 871604, Tempe, Arizona 85287-1604, United States
| | - Jean-Charles Chapuis
- Department of Chemistry and Biochemistry, Arizona State University , P.O. Box 871604, Tempe, Arizona 85287-1604, United States
| |
Collapse
|
19
|
Carral-Menoyo A, Ortiz-de-Elguea V, Martinez-Nunes M, Sotomayor N, Lete E. Palladium-Catalyzed Dehydrogenative Coupling: An Efficient Synthetic Strategy for the Construction of the Quinoline Core. Mar Drugs 2017; 15:md15090276. [PMID: 28867803 PMCID: PMC5618415 DOI: 10.3390/md15090276] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/04/2017] [Accepted: 08/24/2017] [Indexed: 01/20/2023] Open
Abstract
Palladium-catalyzed dehydrogenative coupling is an efficient synthetic strategy for the construction of quinoline scaffolds, a privileged structure and prevalent motif in many natural and biologically active products, in particular in marine alkaloids. Thus, quinolines and 1,2-dihydroquinolines can be selectively obtained in moderate-to-good yields via intramolecular C–H alkenylation reactions, by choosing the reaction conditions. This methodology provides a direct method for the construction of this type of quinoline through an efficient and atom economical procedure, and constitutes significant advance over the existing procedures that require preactivated reaction partners.
Collapse
Affiliation(s)
- Asier Carral-Menoyo
- Departamento de Química Orgánica II, Facultad de Ciencia y Tecnología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.
| | - Verónica Ortiz-de-Elguea
- Departamento de Química Orgánica II, Facultad de Ciencia y Tecnología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.
| | - Mikel Martinez-Nunes
- Departamento de Química Orgánica II, Facultad de Ciencia y Tecnología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.
| | - Nuria Sotomayor
- Departamento de Química Orgánica II, Facultad de Ciencia y Tecnología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.
| | - Esther Lete
- Departamento de Química Orgánica II, Facultad de Ciencia y Tecnología, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.
| |
Collapse
|
20
|
Wang L, Dong C, Li X, Han W, Su X. Anticancer potential of bioactive peptides from animal sources (Review). Oncol Rep 2017; 38:637-651. [PMID: 28677775 DOI: 10.3892/or.2017.5778] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 04/10/2017] [Indexed: 11/06/2022] Open
Abstract
Cancer is the most common cause of human death worldwide. Conventional anticancer therapies, including chemotherapy and radiation, are associated with severe side effects and toxicities as well as low specificity. Peptides are rapidly being developed as potential anticancer agents that specifically target cancer cells and are less toxic to normal tissues, thus making them a better alternative for the prevention and management of cancer. Recent research has focused on anticancer peptides from natural animal sources, such as terrestrial mammals, marine animals, amphibians, and animal venoms. However, the mode of action by which bioactive peptides inhibit the proliferation of cancer cells remains unclear. In this review, we present the animal sources from which bioactive peptides with anticancer activity are derived and discuss multiple proposed mechanisms by which these peptides exert cytotoxic effects against cancer cells.
Collapse
Affiliation(s)
- Linghong Wang
- Clinical Medicine Research Center of the Affiliated Hospital, Inner Mongolia Medical University, Huimin, Hohhot, Inner Mongolia 010050, P.R. China
| | - Chao Dong
- College of Basic Medicine of Inner Mongolia Medical University, Huimin, Hohhot, Inner Mongolia 010050, P.R. China
| | - Xian Li
- Clinical Medicine Research Center of the Affiliated Hospital, Inner Mongolia Medical University, Huimin, Hohhot, Inner Mongolia 010050, P.R. China
| | - Wenyan Han
- Clinical Medicine Research Center of the Affiliated Hospital, Inner Mongolia Medical University, Huimin, Hohhot, Inner Mongolia 010050, P.R. China
| | - Xiulan Su
- Clinical Medicine Research Center of the Affiliated Hospital, Inner Mongolia Medical University, Huimin, Hohhot, Inner Mongolia 010050, P.R. China
| |
Collapse
|
21
|
Newman DJ, Cragg GM. Current Status of Marine-Derived Compounds as Warheads in Anti-Tumor Drug Candidates. Mar Drugs 2017; 15:md15040099. [PMID: 28353637 PMCID: PMC5408245 DOI: 10.3390/md15040099] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 01/09/2023] Open
Abstract
In this review, we have attempted to describe all of the antibody–drug conjugates using a marine-derived compound as the “warhead”, that are currently in clinical trials as listed in the current version of the NIH clinical trials database (clinicaltrials.gov). In searching this database, we used the beta-test version currently available, as it permitted more specific search parameters, since the regular version did not always find trials that had been completed in the past with some agents. We also added small discussion sections on candidates that are still at the preclinical stage, including a derivative of diazonamide that has an unusual interaction with tubulin (DZ-23840), which may also be a potential warhead in the future.
Collapse
|
22
|
Pettit GR, Melody N, Chapuis JC. Antineoplastic Agents. 603. Quinstatins: Exceptional Cancer Cell Growth Inhibitors. JOURNAL OF NATURAL PRODUCTS 2017; 80:692-698. [PMID: 28211277 DOI: 10.1021/acs.jnatprod.6b01006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Discovery of the exceptionally powerful anticancer drug dolastatin 10 (1), contained in the sea hare Dolabella auricularia, opened a new frontier needed for improving human cancer treatment. Subsequently, major advances have been achieved based on results of structurally modifying this unusual natural peptide while maintaining the remarkable anticancer activity necessary for preparation of successful monoclonal antibody drug conjugates (ADC). Among the first several hundred SAR products based on dolastatin 10 our group synthesized and termed auristatins was auristatin E (2a). An anticancer activity-equivalent, desmethylaurisatin E (2b), linked to a CD30 monoclonal antibody is the very successful anticancer drug Adcetris, now approved for use in 65 countries. In the present investigation, we discovered a new subset of auristatins designated quinstatins derived from dolastatin 10 by replacing the C-terminal Doe unit with a carefully designed quinoline, which led to low or subnanomolar levels of cancer cell growth inhibition required for construction of chemically unique ADC drugs. The synthesis of quinstatins 2-8 is presented along with their cancer cell line biological data.
Collapse
Affiliation(s)
- George R Pettit
- Department of Chemistry and Biochemistry, Arizona State University , P.O. Box 871604, Tempe, Arizona 85287-1604, United States
| | - Noeleen Melody
- Department of Chemistry and Biochemistry, Arizona State University , P.O. Box 871604, Tempe, Arizona 85287-1604, United States
| | - Jean-Charles Chapuis
- Department of Chemistry and Biochemistry, Arizona State University , P.O. Box 871604, Tempe, Arizona 85287-1604, United States
| |
Collapse
|
23
|
Mendelsohn BA, Barnscher SD, Snyder JT, An Z, Dodd JM, Dugal-Tessier J. Investigation of Hydrophilic Auristatin Derivatives for Use in Antibody Drug Conjugates. Bioconjug Chem 2017; 28:371-381. [DOI: 10.1021/acs.bioconjchem.6b00530] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Brian A. Mendelsohn
- Agensys Inc. an affiliate
of Astellas Pharma Inc., 1800 Stewart
Street, Santa Monica, California 90404, United States
| | - Stuart D. Barnscher
- Agensys Inc. an affiliate
of Astellas Pharma Inc., 1800 Stewart
Street, Santa Monica, California 90404, United States
| | - Josh T. Snyder
- Agensys Inc. an affiliate
of Astellas Pharma Inc., 1800 Stewart
Street, Santa Monica, California 90404, United States
| | - Zili An
- Agensys Inc. an affiliate
of Astellas Pharma Inc., 1800 Stewart
Street, Santa Monica, California 90404, United States
| | - Jennifer M. Dodd
- Agensys Inc. an affiliate
of Astellas Pharma Inc., 1800 Stewart
Street, Santa Monica, California 90404, United States
| | - Julien Dugal-Tessier
- Agensys Inc. an affiliate
of Astellas Pharma Inc., 1800 Stewart
Street, Santa Monica, California 90404, United States
| |
Collapse
|
24
|
Cyanobacterial Metabolite Calothrixins: Recent Advances in Synthesis and Biological Evaluation. Mar Drugs 2016; 14:17. [PMID: 26771620 PMCID: PMC4728514 DOI: 10.3390/md14010017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/22/2015] [Accepted: 01/04/2016] [Indexed: 12/30/2022] Open
Abstract
The marine environment is host to unparalleled biological and chemical diversity, making it an attractive resource for the discovery of new therapeutics for a plethora of diseases. Compounds that are extracted from cyanobacteria are of special interest due to their unique structural scaffolds and capacity to produce potent pharmaceutical and biotechnological traits. Calothrixins A and B are two cyanobacterial metabolites with a structural assembly of quinoline, quinone, and indole pharmacophores. This review surveys recent advances in the synthesis and evaluation of the biological activities of calothrixins. Due to the low isolation yields from the marine source and the promise this scaffold holds for anticancer and antimicrobial drugs, organic and medicinal chemists around the world have embarked on developing efficient synthetic routes to produce calothrixins. Since the first review appeared in 2009, 11 novel syntheses of calothrixins have been published in the efforts to develop methods that contain fewer steps and higher-yielding reactions. Calothrixins have shown their potential as topoisomerase I poisons for their cytotoxicity in cancer. They have also been observed to target various aspects of RNA synthesis in bacteria. Further investigation into the exact mechanism for their bioactivity is still required for many of its analogs.
Collapse
|
25
|
Maderna A, Leverett CA. Recent advances in the development of new auristatins: structural modifications and application in antibody drug conjugates. Mol Pharm 2015; 12:1798-812. [PMID: 25697404 DOI: 10.1021/mp500762u] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Dolastatin 10 is a powerful antineoplastic agent and microtubule inhibitor that was discovered by Pettit et al. and published in 1987. Since then, many research groups have engaged in SAR studies of synthetic analogues, termed "auristatins". It was eventually discovered that auristatins are of great value as payloads in antibody drug conjugates (ADCs), which led to the FDA-approved ADC brentuximab vedotin (Seattle Genetics). Currently, over 30 ADCs in clinical trials employ auristatins as payloads, and there is a great interest in the research community, both on academic and industrial sides, to further study these analogues. This review will provide an overview of the recent advancements in auristatin development spanning a time frame of about the past ten years. The main focus will be to describe structural changes made to the auristatin peptide and their resulting biological activities in tumor cell proliferation assays. Selected ADC examples will also be described.
Collapse
Affiliation(s)
- Andreas Maderna
- Pfizer Worldwide Research and Development, Worldwide Medicinal Chemistry, Oncology, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Carolyn A Leverett
- Pfizer Worldwide Research and Development, Worldwide Medicinal Chemistry, Oncology, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
26
|
|
27
|
Peng CY, Liu JQ, Zhang R, Shu JC. A new alkaloid from the fruit of Nandina domestica Thunb. Nat Prod Res 2014; 28:1159-64. [DOI: 10.1080/14786419.2014.921166] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Cai-Ying Peng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicines, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P.R. China
| | - Jian-Qun Liu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicines, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P.R. China
| | - Rui Zhang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicines, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P.R. China
| | - Ji-Cheng Shu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicines, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P.R. China
| |
Collapse
|
28
|
Müller P, Martin K, Theurich S, Schreiner J, Savic S, Terszowski G, Lardinois D, Heinzelmann-Schwarz VA, Schlaak M, Kvasnicka HM, Spagnoli G, Dirnhofer S, Speiser DE, von Bergwelt-Baildon M, Zippelius A. Microtubule-depolymerizing agents used in antibody-drug conjugates induce antitumor immunity by stimulation of dendritic cells. Cancer Immunol Res 2014; 2:741-55. [PMID: 24916470 DOI: 10.1158/2326-6066.cir-13-0198] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates (ADC) are emerging as powerful treatment strategies with outstanding target-specificity and high therapeutic activity in patients with cancer. Brentuximab vedotin represents a first-in-class ADC directed against CD30(+) malignancies. We hypothesized that its sustained clinical responses could be related to the stimulation of an anticancer immune response. In this study, we demonstrate that the dolastatin family of microtubule inhibitors, from which the cytotoxic component of brentuximab vedotin is derived, comprises potent inducers of phenotypic and functional dendritic cell (DC) maturation. In addition to the direct cytotoxic effect on tumor cells, dolastatins efficiently promoted antigen uptake and migration of tumor-resident DCs to the tumor-draining lymph nodes. Exposure of murine and human DCs to dolastatins significantly increased their capacity to prime T cells. Underlining the requirement of an intact host immune system for the full therapeutic benefit of dolastatins, the antitumor effect was far less pronounced in immunocompromised mice. We observed substantial therapeutic synergies when combining dolastatins with tumor antigen-specific vaccination or blockade of the PD-1-PD-L1 and CTLA-4 coinhibitory pathways. Ultimately, treatment with ADCs using dolastatins induces DC homing and activates cellular antitumor immune responses in patients. Our data reveal a novel mechanism of action for dolastatins and provide a strong rationale for clinical treatment regimens combining dolastatin-based therapies, such as brentuximab vedotin, with immune-based therapies.
Collapse
Affiliation(s)
- Philipp Müller
- Cancer Immunology & Biology, Department of Biomedicine, Departments of
| | - Kea Martin
- Cancer Immunology & Biology, Department of Biomedicine, Departments of
| | - Sebastian Theurich
- Department I of Internal Medicine and Cologne Interventional Immunology and
| | - Jens Schreiner
- Cancer Immunology & Biology, Department of Biomedicine, Departments of
| | | | | | | | | | - Max Schlaak
- Department of Dermatology, Skin Cancer Center (CIO), University Hospital Cologne, Cologne; and
| | | | - Giulio Spagnoli
- Department of Biomedicine, Institute of Surgical Research and Hospital Management, University of Basel, Basel
| | | | - Daniel E Speiser
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Alfred Zippelius
- Cancer Immunology & Biology, Department of Biomedicine, Departments of Medical Oncology,
| |
Collapse
|
29
|
Telle W, Kelter G, Fiebig HH, Jones PG, Lindel T. Total synthesis and cytotoxicity of the marine natural product malevamide D and a photoreactive analog. Beilstein J Org Chem 2014; 10:316-22. [PMID: 24605152 PMCID: PMC3943603 DOI: 10.3762/bjoc.10.29] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 01/03/2014] [Indexed: 11/23/2022] Open
Abstract
The marine natural product malevamide D from the cyanobacterium Symploca hydnoides was synthesized for the first time. The final peptide coupling linked the dolaisoleuine and dolaproine subunits. The phenyl group of malevamide D was also functionalized with a photoreactive diazirine moiety, which was carried through seven reaction steps. Comprehensive assessment of the cytotoxicity in a panel of 42 human cancer cell lines revealed a geomean IC70 value of 1.5 nM (IC50 0.7 nM) for malevamide D, whereas the photoreactive derivative proved to be less active by a factor of at least 200. COMPARE analysis indicated tubulin interaction as likely mode of action of malevamide D.
Collapse
Affiliation(s)
- Werner Telle
- Institute of Organic Chemistry, TU Braunschweig, Hagenring 30, 38106 Braunschweig, Germany
| | - Gerhard Kelter
- Oncotest Institute for Experimental Oncology GmbH, Am Flughafen 12-14, 79108 Freiburg, Germany
| | - Heinz-Herbert Fiebig
- Oncotest Institute for Experimental Oncology GmbH, Am Flughafen 12-14, 79108 Freiburg, Germany
| | - Peter G Jones
- Institute of Inorganic and Analytical Chemistry, TU Braunschweig, Hagenring 30, 38106 Braunschweig, Germany
| | - Thomas Lindel
- Institute of Organic Chemistry, TU Braunschweig, Hagenring 30, 38106 Braunschweig, Germany
| |
Collapse
|
30
|
Pharmaceutical agents from filamentous marine cyanobacteria. Drug Discov Today 2013; 18:863-71. [DOI: 10.1016/j.drudis.2013.05.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/28/2013] [Accepted: 05/17/2013] [Indexed: 12/30/2022]
|
31
|
Abstract
This review covers the literature published in 2011 for marine natural products, with 870 citations (558 for the period January to December 2011) referring to compounds isolated from marine microorganisms and phytoplankton, green, brown and red algae, sponges, cnidarians, bryozoans, molluscs, tunicates, echinoderms, mangroves and other intertidal plants and microorganisms. The emphasis is on new compounds (1152 for 2011), together with the relevant biological activities, source organisms and country of origin. Biosynthetic studies, first syntheses, and syntheses that lead to the revision of structures or stereochemistries, have been included.
Collapse
Affiliation(s)
- John W Blunt
- Department of Chemistry, University of Canterbury, Christchurch, New Zealand.
| | | | | | | | | |
Collapse
|
32
|
|
33
|
Pettit GR, Rosenberg HJ, Dixon R, Knight JC, Hamel E, Chapuis JC, Pettit RK, Hogan F, Sumner B, Ain KB, Trickey-Platt B. Antineoplastic agents. 548. Synthesis of iodo- and diiodocombstatin phosphate prodrugs. JOURNAL OF NATURAL PRODUCTS 2012; 75:385-93. [PMID: 22324723 PMCID: PMC3313684 DOI: 10.1021/np200797x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Toward the objective of designing a structurally modified analogue of the combretastatin A-4 phosphate prodrug (1b) with the potential for increased specificity toward thyroid carcinoma, synthesis of a series of iodocombstatin phosphate (11a-h) and diiodocombstatin phosphate prodrugs (12a-h) has been accomplished. The diiodo series was obtained via 8a and 9c from condensation of 4 and 6, and the iodo sequence involved a parallel pathway. Both series of iodocombstatins were found to display significant to powerful inhibition of the growth of a panel of human cancer cell lines and of the murine P388 lymphocytic leukemia cell line. Of the diiodo series, 12a was also found to markedly inhibit growth of pediatric neuroblastoma, and monoiodocombstatin 9a strongly inhibited HUVEC growth. Overall, the strongest activity was found against the breast, CNS, leukemia, lung, and prostate cancer cell lines and the least activity against the pancreas and colon lines. Parallel biological investigations of tubulin interaction, antiangiogenesis, and antimicrobial effects were also conducted.
Collapse
Affiliation(s)
- George R Pettit
- Cancer Research Institute and Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona 85287-1604, United States.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|