1
|
Li Y, Bao K, Sun J, Ge R, Zhang Q, Zhang B, Yan X, Li J, Shi F, Zhang M, Zang J, Liu M, Zhou J, Mi W, Xie S, Chen D, Shi L, Dong C. Design of PROTACs utilizing the E3 ligase GID4 for targeted protein degradation. Nat Struct Mol Biol 2025:10.1038/s41594-025-01537-1. [PMID: 40295770 DOI: 10.1038/s41594-025-01537-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/18/2025] [Indexed: 04/30/2025]
Abstract
Proteolysis targeting chimeras (PROTACs) hijack E3 ligases and the ubiquitin-proteasome system to achieve selective degradation of neo-substrates. Their ability to target otherwise intractable substrates has rendered them a valuable modality in drug discovery. However, only a handful of over 600 human E3 ligases have been functionalized for PROTAC applications. Here we show that the E3 ligase GID4 (glucose-induced degradation deficient complex 4) can be leveraged for targeted protein degradation using a noncovalent small molecule. We design and synthesize GID4-based PROTACs, exemplified by NEP162, which can eliminate endogenous BRD4 in a GID4- and ubiquitin-proteasome system-dependent manner. NEP162 exhibits antiproliferative activity and inhibits tumor growth in a xenograft model, hinting toward potential anticancer applications. We further present the crystal structures of GID4-PROTAC-BRD4 ternary complexes in three distinct states, unveiling plastic interactions between GID4 and BRD4. These structural insights, combined with in vitro and in vivo data, decipher the molecular basis by which the hereby developed PROTACs recruit BRD4 to GID4 for targeted degradation and expand our arsenal of PROTAC-exploitable E3 ligases.
Collapse
Affiliation(s)
- Yanran Li
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Kaiwen Bao
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jiyue Sun
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Ruixin Ge
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Qiqing Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Bing Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaojie Yan
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Junlin Li
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Fengying Shi
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Meiling Zhang
- Department of Medicinal Chemistry, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Jinzhi Zang
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Min Liu
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Wenyi Mi
- Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Songbo Xie
- Department of Ophthalmology, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK 'Belt and Road' Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin, China.
| | - Dongxing Chen
- Department of Medicinal Chemistry, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Lei Shi
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| | - Cheng Dong
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, State Key Laboratory of Experimental Hematology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
2
|
Yakkala PA, Kamal A. Dual-targeting inhibitors involving tubulin for the treatment of cancer. Bioorg Chem 2025; 156:108116. [PMID: 39823818 DOI: 10.1016/j.bioorg.2024.108116] [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: 10/05/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/20/2025]
Abstract
Combination therapies play a pivotal role in cancer treatment due to the intricate nature of the disease. Tubulin, a protein crucial for cellular functions, is a prime target in tumor therapy as it regulates microtubule dynamics. Combining tubulin inhibitors with other different inhibitors as dual targeting inhibitors has shown synergistic anti-tumor effects, amplifying therapeutic outcomes. Despite clinical approval of several tubulin inhibitors, their efficacy is hampered by drug resistance and toxic side effects. Dual targeting inhibitors of tubulin and other cancer-related pathways have emerged as vital components in cancer therapy, with promising prospects in both market availability and ongoing clinical trials. The rational design of hybrid inhibitors targeting both pathways presents an innovative approach to combatting cancer. However, despite the potent anti-tumor activity exhibited by several compounds, research on their anti-angiogenic potential remains limited. This review emphasizes the significance of tubulin based dual-target inhibitors, elucidating their mechanisms of action. Recent advances in exploring therapeutic efficacy, toxicity profiles, and challenges such as MDR are discussed. By presenting the research progress of tubulin based dual-target inhibitors as potential anticancer agents, this study delivers valuable insights for the development of more efficient drugs for cancer therapy.
Collapse
Affiliation(s)
- Prasanna Anjaneyulu Yakkala
- Department of Human Nutrition and Analytical Chemistry, Human Nutrition Program, The Ohio State University, Columbus, OH 43212, United States of America; Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ahmed Kamal
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India; Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Dist. Medchal, 500078 TS, India.
| |
Collapse
|
3
|
Wang L, Qi J, Li H, Xu Z. Modular Synthesis of α-Quaternary Chiral β-Lactams via Three-Component Asymmetric Kinugasa/Aldol or Kinugasa/Mannich Cascade Reactions. Chemistry 2025; 31:e202403722. [PMID: 39509604 DOI: 10.1002/chem.202403722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/15/2024]
Abstract
The β-lactam scaffolds are the prevalent structural units in antibiotics and natural products. Herein, three-component asymmetric Kinugasa/aldol and Kinugasa/Mannich cascade reactions have been developed for constructing α-quaternary chiral β-lactams. This method involved the tandem reaction of alkynes, nitrones and aldehydes (or imines), resulting in the formation of three sequential stereocenters controlled by copper(I) catalysts and chiral bis(oxazolidine) ligands. The developed protocol features high enantioselectivity, good reaction efficiency, and mild operational conditions, representing a practical and sustainable synthetic tool for the construction of functionalized chiral β-lactams.
Collapse
Affiliation(s)
- Linxuan Wang
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Jialin Qi
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Haoyu Li
- Dongguan Key Laboratory of Interdisciplinary Science for Advanced Materials and Large-Scale Scientific Facilities, School of Physical Sciences, Great Bay University (Great Bay, Institute for Advanced Study), Dongguan, 523000, China
| | - Zhenghu Xu
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
- State Key Laboratory of Organometallic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| |
Collapse
|
4
|
Ana G, Malebari AM, Noorani S, Fayne D, O’Boyle NM, Zisterer DM, Pimentel EF, Endringer DC, Meegan MJ. ( E)-1-(3-(3-Hydroxy-4-Methoxyphenyl)-1-(3,4,5-Trimethoxyphenyl)allyl)-1 H-1,2,4-Triazole and Related Compounds: Their Synthesis and Biological Evaluation as Novel Antimitotic Agents Targeting Breast Cancer. Pharmaceuticals (Basel) 2025; 18:118. [PMID: 39861179 PMCID: PMC11769294 DOI: 10.3390/ph18010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: The synthesis of (E)-1-(1,3-diphenylallyl)-1H-1,2,4-triazoles and related compounds as anti-mitotic agents with activity in breast cancer was investigated. These compounds were designed as hybrids of the microtubule-targeting chalcones, indanones, and the aromatase inhibitor letrozole. Methods: A panel of 29 compounds was synthesized and examined by a preliminary screening in estrogen receptor (ER) and progesterone receptor (PR)-positive MCF-7 breast cancer cells together with cell cycle analysis and tubulin polymerization inhibition. Results: (E)-5-(3-(1H-1,2,4-triazol-1-yl)-3-(3,4,5-trimethoxyphenyl)prop-1-en-1-yl)-2-methoxyphenol 22b was identified as a potent antiproliferative compound with an IC50 value of 0.39 mM in MCF-7 breast cancer cells, 0.77 mM in triple-negative MDA-MB-231 breast cancer cells, and 0.37 mM in leukemia HL-60 cells. In addition, compound 22b demonstrated potent activity in the sub-micromolar range against the NCI 60 cancer cell line panel including prostate, melanoma, colon, leukemia, and non-small cell lung cancers. G2/M phase cell cycle arrest and the induction of apoptosis in MCF-7 cells together with inhibition of tubulin polymerization were demonstrated. Immunofluorescence studies confirmed that compound 22b targeted tubulin in MCF-7 cells, while computational docking studies predicted binding conformations for 22b in the colchicine binding site of tubulin. Compound 22b also selectively inhibited aromatase. Conclusions: Based on the results obtained, these novel compounds are suitable candidates for further investigation as antiproliferative microtubule-targeting agents for breast cancer.
Collapse
Affiliation(s)
- Gloria Ana
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Azizah M. Malebari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sara Noorani
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Darren Fayne
- Molecular Design Group, School of Chemical Sciences, Dublin City University, Glasnevin, D09 V209 Dublin, Ireland
- DCU Life Sciences Institute, Dublin City University, Glasnevin, D09 V209 Dublin, Ireland
| | - Niamh M. O’Boyle
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Daniela M. Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, D02 R590 Dublin, Ireland
| | - Elisangela Flavia Pimentel
- Department of Pharmaceutical Sciences, University Vila Velha, Av. Comissário José Dantas de Melo, n°21, Boa Vista, Vila Velha CEP 29102-920, Brazil
| | - Denise Coutinho Endringer
- Department of Pharmaceutical Sciences, University Vila Velha, Av. Comissário José Dantas de Melo, n°21, Boa Vista, Vila Velha CEP 29102-920, Brazil
| | - Mary J. Meegan
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| |
Collapse
|
5
|
Xiong S, Song K, Xiang H, Luo G. Dual-target inhibitors based on ERα: Novel therapeutic approaches for endocrine resistant breast cancer. Eur J Med Chem 2024; 270:116393. [PMID: 38588626 DOI: 10.1016/j.ejmech.2024.116393] [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: 05/25/2023] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024]
Abstract
Estrogen receptor alpha (ERα), a nuclear transcription factor, is a well-validated therapeutic target for more than 70% of all breast cancers (BCs). Antagonizing ERα either by selective estrogen receptor modulators (SERMs) or selective estrogen receptor degraders (SERDs) forms the foundation of endocrine therapy and has achieved great success in the treatment of ERα positive (ERα+) BCs. Unfortunately, despite initial effectiveness, endocrine resistance eventually emerges in up to 30% of ERα+ BC patients and remains a significant medical challenge. Several mechanisms implicated in endocrine resistance have been extensively studied, including aberrantly activated growth factor receptors and downstream signaling pathways. Hence, the crosstalk between ERα and another oncogenic signaling has led to surge of interest to develop combination therapies and dual-target single agents. This review briefly introduces the synergisms between ERα and another anticancer target and summarizes the recent advances of ERα-based dual-targeting inhibitors from a medicinal chemistry perspective. Accordingly, their rational design strategies, structure-activity relationships (SARs) and biological activities are also dissected to provide some perspectives on future directions for ERα-based dual target drug discovery in BC therapy.
Collapse
Affiliation(s)
- Shuangshuang Xiong
- Jiangsu Key Laboratory of Drug Design and Optimization, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ke Song
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hua Xiang
- Jiangsu Key Laboratory of Drug Design and Optimization, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Guoshun Luo
- Jiangsu Key Laboratory of Drug Design and Optimization, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
6
|
Kurian J, Ashtam A, Kesavan A, Chaluvalappil SV, Panda D, Manheri MK. Hybridization of the Pharmacophoric Features of Discoipyrrole C and Combretastatin A-4 toward New Anticancer Leads. ChemMedChem 2023; 18:e202300081. [PMID: 37256820 DOI: 10.1002/cmdc.202300081] [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: 02/12/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/02/2023]
Abstract
Pharmacophore hybridization is an attractive strategy to identify new leads against multifactorial diseases such as cancer. Based on literature analysis of compounds possessing 'vicinal diaryl' fragment in their structure, we considered Discoipyrroles A-D and Combretastatin A-4 (CA-4) as possible components in hybrid design. Discoipyrrole C (Dis C) and CA-4 were used as reference compounds in these studies and their hybrids, in the form of 4,5-diaryl-1H-pyrrol-3(2H)-ones, were synthesized from suitable amino acid precursors though their ynone intermediates. Of these, the hybrid having exact substitution pattern as that of CA-4 showed better potency and selectivity than Dis C, but its activity was less compared to CA-4. This new analog disrupted interphase microtubules by inhibiting tubulin assembly by binding to the colchicine site, induced multipolar spindles, caused cell cycle block and apoptosis in HeLa cells. It also inhibited colony formation and migration of breast cancer cell lines.
Collapse
Affiliation(s)
- Jais Kurian
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, 600036, Tamil Nadu, India
| | - Anvesh Ashtam
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Akila Kesavan
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, 600036, Tamil Nadu, India
| | | | - Dulal Panda
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
- National Institute of Pharmaceutical Education and Research, SAS Nagar, Punjab, 160062, India
| | - Muraleedharan K Manheri
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, 600036, Tamil Nadu, India
| |
Collapse
|
7
|
Xin L, Min J, Hu H, Li Y, Du C, Xie B, Cheng Y, Deng X, Deng X, Shen K, Huang J, Chen CC, Guo RT, Dong C, Zhou HB. Structure-guided identification of novel dual-targeting estrogen receptor α degraders with aromatase inhibitory activity for the treatment of endocrine-resistant breast cancer. Eur J Med Chem 2023; 253:115328. [PMID: 37037140 DOI: 10.1016/j.ejmech.2023.115328] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/12/2023]
Abstract
Drug resistance is a major challenge in conventional endocrine therapy for estrogen receptor (ER) positive breast cancer (BC). BC is a multifactorial disease, in which simultaneous aromatase (ARO) inhibition and ERα degradation may effectively inhibit the signal transduction of both proteins, thus potentially overcoming drug resistance caused by overexpression or mutation of target proteins. In this study, guided by the X-ray structure of a hit compound 30a in complex with ER-Y537S, a structure-based optimization was performed to get a series of multiacting inhibitors targeting both ERα and ARO, and finally a novel class of potent selective estrogen receptor degraders (SERDs) based on a three-dimensional oxabicycloheptene sulfonamide (OBHSA) scaffold equipped with aromatase inhibitor (AI) activity were identified. Of these dual-targeting SERD-AI hybrids, compound 31q incorporating a 1H-1,2,4-triazole moiety showed excellent ERα degradation activity, ARO inhibitory activity and remarkable antiproliferative activity against BC resistant cells. Furthermore, 31q manifested efficient tumor suppression in MCF-7 tumor xenograft models. Taken together, our study reported for the first time the highly efficient dual-targeting SERD-AI hybrid compounds, which may lay the foundation of translational research for improved treatment of endocrine-resistant BC.
Collapse
Affiliation(s)
- Lilan Xin
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jian Min
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Hebing Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Yuanyuan Li
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Chuanqian Du
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Baohua Xie
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yan Cheng
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiaofei Deng
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiangping Deng
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Kang Shen
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jian Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Chun-Chi Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Rey-Ting Guo
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China.
| | - Chune Dong
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Hai-Bing Zhou
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; Frontier Science Center for Immunology and Metabolism, State Key Laboratory of Virology, Provincial Key Laboratory of Developmentally Originated Disease, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
8
|
Li N, Guan Q, Hong Y, Zhang B, Li M, Li X, Li B, Wu L, Zhang W. Discovery of 6-aryl-2-(3,4,5-trimethoxyphenyl)thiazole[3,2-b][1,2,4]triazoles as potent tubulin polymerization inhibitors. Eur J Med Chem 2023; 256:115402. [PMID: 37182330 DOI: 10.1016/j.ejmech.2023.115402] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/16/2023]
Abstract
Tubulin/colchicine-binding site inhibitors (CBSIs) co-crystal structures play an important role in the exploration of novel small molecules for oncotherapy. Based on the analysis of the binding models of tubulin and reported CBSIs, a series of 6-aryl-2-(3,4,5-trimethoxyphenyl)thiazole[3,2-b][1,2,4]triazoles were designed as potential tubulin polymerization inhibitors by binding to distinct colchicine domain of tubulin. Among the compounds synthesized, 7w not only shown noteworthy potency against SGC-7901 cancer cell line (IC50 = 0.21 μM) but also exhibited lower cytotoxicity than colchicine in normal cell line (HUVEC). The mechanism studies elucidated that 7w could cause the apoptosis of cancer cells by inhibiting tubulin polymerization to trigger G2/M arrest. In 4T1-xenograft mice model, 7w significantly inhibited tumor growth without losing weight, demonstrating a promising potential for further development with a unique binding mode at the colchicine-binding site.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Yilang Hong
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Bowen Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Mi Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xuewen Li
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Street, Heping District, Shenyang, 110002, China
| | - Bo Li
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Street, Heping District, Shenyang, 110002, China.
| | - Lan Wu
- Department of Geratology, The First Affiliated Hospital, Chinese Medical University, Shenyang, 110001, China.
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
9
|
Hawash M. Recent Advances of Tubulin Inhibitors Targeting the Colchicine Binding Site for Cancer Therapy. Biomolecules 2022; 12:biom12121843. [PMID: 36551271 PMCID: PMC9776383 DOI: 10.3390/biom12121843] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer accounts for numerous deaths each year, and it is one of the most common causes of death worldwide, despite many breakthroughs in the discovery of novel anticancer candidates. Each new year the FDA approves the use of new drugs for cancer treatments. In the last years, the biological targets of anticancer agents have started to be clearer and one of these main targets is tubulin protein; this protein plays an essential role in cell division, as well as in intracellular transportation. The inhibition of microtubule formation by targeting tubulin protein induces cell death by apoptosis. In the last years, numerous novel structures were designed and synthesized to target tubulin, and this can be achieved by inhibiting the polymerization or depolymerization of the microtubules. In this review article, recent novel compounds that have antiproliferation activities against a panel of cancer cell lines that target tubulin are explored in detail. This review article emphasizes the recent developments of tubulin inhibitors, with insights into their antiproliferative and anti-tubulin activities. A full literature review shows that tubulin inhibitors are associated with properties in the inhibition of cancer cell line viability, inducing apoptosis, and good binding interaction with the colchicine binding site of tubulin. Furthermore, some drugs, such as cabazitaxel and fosbretabulin, have been approved by FDA in the last three years as tubulin inhibitors. The design and development of efficient tubulin inhibitors is progressively becoming a credible solution in treating many species of cancers.
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine
| |
Collapse
|
10
|
Discovery of N-benzylarylamide derivatives as novel tubulin polymerization inhibitors capable of activating the Hippo pathway. Eur J Med Chem 2022; 240:114583. [PMID: 35834904 DOI: 10.1016/j.ejmech.2022.114583] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 01/12/2023]
Abstract
Novel N-benzylarylamide saderivatives were designed and synthesized, and their antiproliferative activities were explored. Some of 51 target compounds exhibited potent inhibitory activities against MGC-803, HCT-116 and KYSE450 cells with IC50 values in two-digit nanomolar. Compound I-33 (MY-875) displayed the most potent antiproliferative activities against MGC-803, HCT-116 and KYSE450 cells (IC50 = 0.027, 0.055 and 0.067 μM, respectively) and possessed IC50 values ranging from 0.025 to 0.094 μM against other 11 cancer cell lines. Further mechanism studies indicated that compound I-33 (MY-875) inhibited tubulin polymerization (IC50 = 0.92 μM) by targeting the colchicine bingding site of tubulin. Compound I-33 (MY-875) disrupted the construction of the microtubule networks and affected the mitosis in MGC-803 and SGC-7901 cells. In addition, although it acted as a colchicine binding site inhibitor, compound I-33 (MY-875) also activated the Hippo pathway to promote the phosphorylation status of MST and LATS, resulting in the YAP degradation in MGC-803 and SGC-7901 cells. Due to the degradation of YAP, the expression levels of TAZ and Axl decreased. Because of the dual actions on colchicine binding site and Hippo pathway, compound I-33 (MY-875) dose-dependently inhibited cell colony formatting ability, arrested cells at the G2/M phase and induced cells apoptosis in MGC-803 and SGC-7901 cells. Moreover, compound I-33 (MY-875) could regulate the levels of cell cycle and apoptosis regulatory proteins in MGC-803 and SGC-7901 cells. Furthermore, molecular docking analysis suggested that the hydrogen bond and hydrophobic interactions made compound I-33 (MY-875) well bind into the colchicine binding site of tubulin. Collectively, compound I-33 (MY-875) is a novel anti-gastric cancer agent and deserves to be further investigated for cancer therapy by targeting the colchicine binding site of tubulin and activating the Hippo pathway.
Collapse
|
11
|
A novel aromatic amide derivative SY-65 co-targeted tubulin and histone deacetylase 1 with potent anticancer activity in vitro and in vivo. Biochem Pharmacol 2022; 201:115070. [DOI: 10.1016/j.bcp.2022.115070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/27/2022]
|
12
|
A potent estrogen receptor and microtubule specific purine-benzothiazole-based fluorescent molecular probe induces apoptotic death of breast cancer cells. Sci Rep 2022; 12:10772. [PMID: 35750870 PMCID: PMC9232585 DOI: 10.1038/s41598-022-12933-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is the most common malignancy in women and is a heterogeneous disease at molecular level. Early detection and specificity are the key prerequisite for the treatment of this deadly cancer. To address these issues attention on the breast cancer specific receptor protein(s) is the most realistic option. Herein estrogen (E) and progesterone (Pg) receptors(R) were considered to design fluorescent molecular probes with possible therapeutic option. We adopted QSAR technique to design a library of benzothiazole-purine hybrid molecules. Molecular docking offers us three screened molecules as most potential. Among these molecules one abbreviated as “CPIB” showed blue fluorescence and detected ER positive cancer cells at 1 nM concentration. At elevated concentration, CPIB induces apoptotic deaths of same cancer cells through targeting intracellular microtubules without affecting normal cells or ER negative cells. CPIB is one of its kind with two-in-one potential of “Detection and Destroy” ability targeting ER positive breast cancer cells.
Collapse
|
13
|
Catalyst-controlled regiodivergent 1,2-difunctionalization of alkenes with two carbon-based electrophiles. Sci China Chem 2021. [DOI: 10.1007/s11426-021-1172-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
14
|
Discovery of Novel Diarylamide N-Containing Heterocyclic Derivatives as New Tubulin Polymerization Inhibitors with Anti-Cancer Activity. Molecules 2021; 26:molecules26134047. [PMID: 34279387 PMCID: PMC8272053 DOI: 10.3390/molecules26134047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 01/11/2023] Open
Abstract
Tubulin has been regarded as an attractive and successful molecular target in cancer therapy and drug discovery. Vicinal diaryl is a simple scaffold found in many colchicine site tubulin inhibitors, which is also an important pharmacophoric point of tubulin binding and anti-cancer activity. As the continuation of our research work on colchicine binding site tubulin inhibitors, we designed and synthesized a series of diarylamide N-containing heterocyclic derivatives by the combination of vicinal diaryl core and N-containing heterocyclic skeletons into one hybrid though proper linkers. Among of these compounds, compound 15b containing a 5-methoxyindole group exhibited the most potent inhibitory activity against the tested three human cancer cell lines (MGC-803, PC-3 and EC-109) with IC50 values of 1.56 μM, 3.56 μM and 14.5 μM, respectively. Besides, the SARs of these compounds were preliminarily studied and summarized. The most active compound 15b produced the inhibition of tubulin polymerization in a dose-dependent manner and caused microtubule network disruption in MGC-803 cells. Therefore, compound 15b was identified as a novel tubulin polymerization inhibitor targeting the colchicine binding site. In addition, the results of molecular docking also suggested compound 15b could tightly bind into the colchicine binding site of β-tubulin.
Collapse
|
15
|
Shuai W, Wang G, Zhang Y, Bu F, Zhang S, Miller DD, Li W, Ouyang L, Wang Y. Recent Progress on Tubulin Inhibitors with Dual Targeting Capabilities for Cancer Therapy. J Med Chem 2021; 64:7963-7990. [PMID: 34101463 DOI: 10.1021/acs.jmedchem.1c00100] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Microtubules play a crucial role in multiple cellular functions including mitosis, cell signaling, and organelle trafficking, which makes the microtubule an important target for cancer therapy. Despite the great successes of microtubule-targeting agents in the clinic, the development of drug resistance and dose-limiting toxicity restrict their clinical efficacy. In recent years, multitarget therapy has been considered an effective strategy to achieve higher therapeutic efficacy, in particular dual-target drugs. In terms of the synergetic effect of tubulin and other antitumor agents such as receptor tyrosine kinases inhibitors, histone deacetylases inhibitors, DNA-damaging agents, and topoisomerase inhibitors in combination therapy, designing dual-target tubulin inhibitors is regarded as a promising approach to overcome these limitations and improve therapeutic efficacy. In this Perspective, we discussed rational target combinations, design strategies, structure-activity relationships, and future directions of dual-target tubulin inhibitors.
Collapse
Affiliation(s)
- Wen Shuai
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yiwen Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Faqian Bu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Sicheng Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, Innovation Center of Nursing Research, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
16
|
Ana G, Kelly PM, Malebari AM, Noorani S, Nathwani SM, Twamley B, Fayne D, O’Boyle NM, Zisterer DM, Pimentel EF, Endringer DC, Meegan MJ. Synthesis and Biological Evaluation of 1-(Diarylmethyl)-1 H-1,2,4-triazoles and 1-(Diarylmethyl)-1 H-imidazoles as a Novel Class of Anti-Mitotic Agent for Activity in Breast Cancer. Pharmaceuticals (Basel) 2021; 14:169. [PMID: 33671674 PMCID: PMC7926793 DOI: 10.3390/ph14020169] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/21/2022] Open
Abstract
We report the synthesis and biochemical evaluation of compounds that are designed as hybrids of the microtubule targeting benzophenone phenstatin and the aromatase inhibitor letrozole. A preliminary screening in estrogen receptor (ER)-positive MCF-7 breast cancer cells identified 5-((2H-1,2,3-triazol-1-yl)(3,4,5-trimethoxyphenyl)methyl)-2-methoxyphenol 24 as a potent antiproliferative compound with an IC50 value of 52 nM in MCF-7 breast cancer cells (ER+/PR+) and 74 nM in triple-negative MDA-MB-231 breast cancer cells. The compounds demonstrated significant G2/M phase cell cycle arrest and induction of apoptosis in the MCF-7 cell line, inhibited tubulin polymerisation, and were selective for cancer cells when evaluated in non-tumorigenic MCF-10A breast cells. The immunofluorescence staining of MCF-7 cells confirmed that the compounds targeted tubulin and induced multinucleation, which is a recognised sign of mitotic catastrophe. Computational docking studies of compounds 19e, 21l, and 24 in the colchicine binding site of tubulin indicated potential binding conformations for the compounds. Compounds 19e and 21l were also shown to selectively inhibit aromatase. These compounds are promising candidates for development as antiproliferative, aromatase inhibitory, and microtubule-disrupting agents for breast cancer.
Collapse
Affiliation(s)
- Gloria Ana
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Patrick M. Kelly
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Azizah M. Malebari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Sara Noorani
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Seema M. Nathwani
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.F.); (D.M.Z.)
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, DO2R590 Dublin, Ireland;
| | - Darren Fayne
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.F.); (D.M.Z.)
| | - Niamh M. O’Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Daniela M. Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.F.); (D.M.Z.)
| | - Elisangela Flavia Pimentel
- Department of Pharmaceutical Sciences, University Vila Velha, Av. Comissário José Dantas de Melo, n°21, Boa Vista Vila Velha—Espírito Santo, Vila Velha 29102-920, Brazil; (E.F.P.); (D.C.E.)
| | - Denise Coutinho Endringer
- Department of Pharmaceutical Sciences, University Vila Velha, Av. Comissário José Dantas de Melo, n°21, Boa Vista Vila Velha—Espírito Santo, Vila Velha 29102-920, Brazil; (E.F.P.); (D.C.E.)
| | - Mary J. Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| |
Collapse
|
17
|
Lin S, Liang Y, Cheng J, Pan F, Wang Y. Novel diaryl-2H-azirines: Antitumor hybrids for dual-targeting tubulin and DNA. Eur J Med Chem 2021; 214:113256. [PMID: 33581556 DOI: 10.1016/j.ejmech.2021.113256] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 01/30/2023]
Abstract
Multiple-target drugs may achieve better therapeutic effect via different pathways than single-target ones, especially for complex diseases. Tubulin and DNA are well-characterized molecular targets for anti-cancer drug development. A novel class of diaryl substituted 2H-azirines were designed based on combination of pharmacophores from Combretastatin A-4 (CA-4) and aziridine-type alkylating agents, which are known tubulin polymerization inhibitor and DNA damaging agents, respectively. The antitumor activities of these compounds were evaluated in vitro and 6h showed the most potent activities against four cancer cell lines with IC50 values ranging from 0.16 to 1.40 μM. Further mechanistic studies revealed that 6h worked as a bifunctional agent targeting both tubulin and DNA. In the nude mice xenograft model, 6h significantly inhibited the tumor growth with low toxicity, demonstrating the promising potential for further developing novel cancer therapy with a unique mechanism.
Collapse
Affiliation(s)
- Shibo Lin
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yuru Liang
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jiayi Cheng
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Feng Pan
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yang Wang
- School of Pharmacy, Fudan University, Shanghai, 201203, China; Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
18
|
Thakur S, Das A, Das T. 1,3-Dipolar cycloaddition of nitrones: synthesis of multisubstituted, diverse range of heterocyclic compounds. NEW J CHEM 2021. [DOI: 10.1039/d1nj02023d] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The 1,3-dipolar cycloaddition reaction of nitrone is one of the most important methods for the synthesis of different sizes of heterocycles which have enormous applications in natural products, biologically active molecules and pharmaceuticals.
Collapse
Affiliation(s)
- Seema Thakur
- Department of Chemistry
- NIT Jamshedpur
- Jamshedpur 831014
- India
| | - Arunima Das
- Department of Chemistry
- NIT Jamshedpur
- Jamshedpur 831014
- India
| | - Tapas Das
- Department of Chemistry
- NIT Jamshedpur
- Jamshedpur 831014
- India
| |
Collapse
|
19
|
Twamley B, O’Boyle NM, Meegan MJ. Azetidin-2-ones: structures of anti-mitotic compounds based on the 1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one core. Acta Crystallogr E Crystallogr Commun 2020; 76:1187-1194. [PMID: 32843997 PMCID: PMC7405576 DOI: 10.1107/s2056989020008555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 11/15/2022]
Abstract
A series of related substituted 1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-ones have been characterized: 3-(4-fluoro-phen-yl)-4-(4-meth-oxy-phen-yl)-1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one, C25H24FNO5 (1), 3-(furan-2-yl)-4-(4-meth-oxy-phen-yl)-1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one, C23H23NO6 (2), 4-(4-meth-oxyphen-yl)-3-(naphthalen-1-yl)-1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one, C29H27NO5 (3), 3-(3,4-di-meth-oxy-phen-yl)-4-(4-meth-oxy-phen-yl)-1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one, C27H29NO7 (4) and 4,4-bis-(4-meth-oxy-phen-yl)-3-phenyl-1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one, C32H31NO6 (5). All of the compounds are racemic. The lactam and 3,4,5-tri-meth-oxy-phenyl rings are approximately co-planar and the orientation of the lactam and the 4-meth-oxy-phenyl substituent is approximately orthogonal. The chiral centres, although eclipsed by geometry, have torsion angles ranging from -7.27 to 13.08° for the 3 position, and -8.69 to 13.76° for the 4 position of the β-lactam. The structures display intra-molecular C-H⋯O bonding between the 3,4,5-tri-meth-oxy-phenyl ring and the lactam ketone. Further C-H⋯O inter-actions are observed and form either an opposing meth-oxy 'buckle' to join two mol-ecules together or a cyclic dimer.
Collapse
Affiliation(s)
- Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Niamh M. O’Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152 - 160 Pearse St, Dublin 2, Ireland
| | - Mary J. Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152 - 160 Pearse St, Dublin 2, Ireland
| |
Collapse
|
20
|
Fang Y, Liao G, Guo H, Yu B, Liu HM. Synthesis of biologically relevant steroidal spiro β-lactams from dienamides through the cascade 4-endo N-cyclization/aerobic oxidation sequence. Steroids 2020; 159:108635. [PMID: 32169578 DOI: 10.1016/j.steroids.2020.108635] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/27/2020] [Accepted: 03/04/2020] [Indexed: 11/26/2022]
Abstract
The steroid nucleus and β-lactam are prevalent in natural products and drug molecules, the compounds containing such fragments always possess diverse and interesting biological profiles. Presented here is an unprecedented cascade 4-endo N-cyclization/aerobic oxidation sequence that enables the synthesis of biologically relevant steroidal spiro β-lactams from dienamides. Of note, two continuous quaternary chiral centers were constructed simultaneously in this process, and the title compounds bearing the OH and CN groups are highly functionalized, allowing for late-stage transformations for construction of diverse compound library. The protocol has several advantages such as mild reaction conditions and short reaction time, therefore could serve as a new strategy for synthesizing β-lactams.
Collapse
Affiliation(s)
- Yuan Fang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Guochao Liao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Hao Guo
- Chongqing Institute of Forensic Science, Chongqing 400021, China
| | - Bin Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
21
|
Fu DJ, Zhang YF, Chang AQ, Li J. β-Lactams as promising anticancer agents: Molecular hybrids, structure activity relationships and potential targets. Eur J Med Chem 2020; 201:112510. [PMID: 32592915 DOI: 10.1016/j.ejmech.2020.112510] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 01/17/2023]
Abstract
β-Lactam, commonly referred as azetidin-2-one, is a multifunctional building block for synthesizing β-amino ketones, γ-amino alcohols, and other compounds. Besides its well known antibiotic activity, this ring system exhibits a wide range of activities, attracting the attention of researchers. However, the structurally diverse β-lactam analogues as anticancer agents and their different molecular targets are poorly discussed. The purpose of this review is 3-fold: (1) to explore the molecular hybridization approach to design β-lactams hybrids as anticancer agents; (2) the structure activity relationship of the most active anticancer β-lactams and (3) to summarize their antitumor mechanisms.
Collapse
Affiliation(s)
- Dong-Jun Fu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Yun-Feng Zhang
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - An-Qi Chang
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Jun Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China.
| |
Collapse
|
22
|
da Silveira Pinto LS, Vasconcelos TRA, Gomes CRB, de Souza MVN. A Brief Review on the Development of Novel Potentially Active Azetidin-2-ones Against Cancer. CURR ORG CHEM 2020. [DOI: 10.2174/1385272824666200303115444] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Azetidin-2-ones (β-lactams) and its derivatives are an important group of heterocyclic compounds that exhibit a wide range of pharmacological properties such as antibacterial, anticancer, anti-diabetic, anti-inflammatory and anticonvulsant. Efforts have been made over the years to develop novel congeners with superior biological activities and minimal potential for undesirable side effects. The present review aimed to highlight some recent discoveries (2013-2019) on the development of novel azetidin-2-one-based compounds as potential anticancer agents.
Collapse
Affiliation(s)
- Ligia S. da Silveira Pinto
- Universidade Federal Fluminense, Instituto de Quimica, Departamento de Quimica Organica, Programa de Pos-Graduacao em Quimica. Outeiro de Sao Joao Batista, s/no, Centro, Niteroi, 24020-141, Rio de Janeiro, Brazil
| | - Thatyana R. Alves Vasconcelos
- Universidade Federal Fluminense, Instituto de Quimica, Departamento de Quimica Organica, Programa de Pos-Graduacao em Quimica. Outeiro de Sao Joao Batista, s/no, Centro, Niteroi, 24020-141, Rio de Janeiro, Brazil
| | - Claudia Regina B. Gomes
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos-Farmanguinhos. Rua Sizenando Nabuco 100, Manguinhos, 21041-250, Rio de Janeiro, RJ, Brazil
| | - Marcus Vinícius N. de Souza
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos-Farmanguinhos. Rua Sizenando Nabuco 100, Manguinhos, 21041-250, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
23
|
Zhang X, Jia Y. Recent Advances in β-lactam Derivatives as Potential Anticancer Agents. Curr Top Med Chem 2020; 20:1468-1480. [PMID: 32148196 DOI: 10.2174/1568026620666200309161444] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 01/26/2023]
Abstract
Cancer, accounts for around 10 million deaths annually, is the second leading cause of death globally. The continuous emergency of drug-resistant cancers and the low specificity of anticancer agents are the main challenges in the control and eradication of cancers, so it is imperative to develop novel anticancer agents. Immense efforts have been made in developing new lead compounds and novel chemotherapeutic strategies for the treatment of various forms of cancers in recent years. β-Lactam derivatives constitute versatile and attractive scaffolds for the drug discovery since these kinds of compounds possess a variety of pharmacological properties, and some of them exhibited promising potency against both drug-sensitive and drug-resistant cancer cell lines. Thus, β-lactam moiety is a useful template for the development of novel anticancer agents. This review will provide an overview of β-lactam derivatives with the potential therapeutic application for the treatment of cancers covering articles published between 2000 and 2020. The mechanisms of action, the critical aspects of design and structureactivity relationships are also discussed.
Collapse
Affiliation(s)
- Xinfen Zhang
- Department of Oncology, Zhuji Affiliated Hospital of Shaoxing University, Zhejiang Province 311800, China
| | - Yanshu Jia
- Chongqing Institute of Engineering, Chongqing 400056, China
| |
Collapse
|
24
|
Makar S, Saha T, Swetha R, Gutti G, Kumar A, Singh SK. Rational approaches of drug design for the development of selective estrogen receptor modulators (SERMs), implicated in breast cancer. Bioorg Chem 2019; 94:103380. [PMID: 31757413 DOI: 10.1016/j.bioorg.2019.103380] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/20/2022]
Abstract
Drug discovery and development have gained momentum due to the rational drug design by engaging computational tools and bioinformatics methodologies. Bioisosteric replacements and hybrid molecular approaches are the other inventive processes, used by medicinal chemists for the desired modifications of leads for clinical drug candidates. SERMs, ought to produce inhibitory activity in breast, uterus and agonist activity in other tissues, are beneficial for estrogen-like actions. ER subtypes α and β are hormone dependent modulators of intracellular signaling and gene expression, and development of ER selective ligands could be an effective approach for treatment of breast cancer. This report has critically investigated the possible designing considerations of SERMs, their in silico interactions, and potent pharmacophore generation approaches viz. indole, restricted benzothiophene [3, 2-b] indole, carborane, xanthendione, combretastatin A-4, organometallic heterocycles, OBHS-SAHA hybrids, benzopyranones, tetrahydroisoquinolines, Dig G derivatives and their specifications in drug design and development, to rationally improve the understanding in drug discovery. This also includes various strategies for the development of dual inhibitors for the management of antiestrogenic resistance.
Collapse
Affiliation(s)
- Subhajit Makar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India
| | - Tanmay Saha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India
| | - Rayala Swetha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India
| | - Gopichand Gutti
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India
| | - Sushil K Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P, India.
| |
Collapse
|
25
|
Duan Y, Liu W, Tian L, Mao Y, Song C. Targeting Tubulin-colchicine Site for Cancer Therapy: Inhibitors, Antibody- Drug Conjugates and Degradation Agents. Curr Top Med Chem 2019; 19:1289-1304. [PMID: 31210108 DOI: 10.2174/1568026619666190618130008] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/22/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022]
Abstract
Microtubules are essential for the mitotic division of cells and have been an attractive target
for antitumour drugs due to the increased incidence of cancer and significant mitosis rate of tumour cells.
In the past few years, tubulin-colchicine binding site, as one of the three binding pockets including taxol-,
vinblastine- and colchicine-binding sites, has been focused on to design tubulin-destabilizing agents including
inhibitors, antibody-drug conjugates and degradation agents. The present review is the first to
cover a systemic and recent synopsis of tubulin-colchicine binding site agents. We believe that it would
provide an increase in our understanding of receptor-ligand interaction pattern and consciousness of a
series of challenges about tubulin target druggability.
Collapse
Affiliation(s)
- Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Wei Liu
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Liang Tian
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Yanna Mao
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Chuanjun Song
- College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
26
|
Wei C, Zhu J, Zhang J, Deng Q, Mo D. Synthesis of Spirofluorenyl‐
β
‐Lactams through Cycloaddition and Ring Contraction from
N
‐Aryl Fluorenone Nitrones and Methylenecyclopropanes. Adv Synth Catal 2019. [DOI: 10.1002/adsc.201900523] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Cui Wei
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China; School of Chemistry and Pharmaceutical Sciences Guangxi Normal University 15 Yu Cai Road Guilin 541004 People's Republic of China
| | - Jie‐Feng Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China; School of Chemistry and Pharmaceutical Sciences Guangxi Normal University 15 Yu Cai Road Guilin 541004 People's Republic of China
| | - Jin‐Qi Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China; School of Chemistry and Pharmaceutical Sciences Guangxi Normal University 15 Yu Cai Road Guilin 541004 People's Republic of China
| | - Qi Deng
- School of Chemistry and Chemical Engineering Hunan University of Science and Technology Xiangtan 411201 People's Republic of China
| | - Dong‐Liang Mo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China; School of Chemistry and Pharmaceutical Sciences Guangxi Normal University 15 Yu Cai Road Guilin 541004 People's Republic of China
| |
Collapse
|
27
|
Hosseini A, Schreiner PR. Synthesis of Exclusively 4-Substituted β-Lactams through the Kinugasa Reaction Utilizing Calcium Carbide. Org Lett 2019; 21:3746-3749. [PMID: 31059273 DOI: 10.1021/acs.orglett.9b01192] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A new Kinugasa reaction protocol has been elaborated for the one-pot synthesis of 4-substituted β-lactams utilizing calcium carbide and nitrone derivatives. Calcium carbide is thereby activated by TBAF·3H2O in the presence of CuCl/NMI. The ease of synthesis and use of inexpensive chemicals provides rapid access of practical quantities of β-lactams exclusively substituted at position 4.
Collapse
Affiliation(s)
- Abolfazl Hosseini
- Institute of Organic Chemistry , Justus Liebig University , Heinrich-Buff-Ring 17 , 35392 Giessen , Germany
| | - Peter R Schreiner
- Institute of Organic Chemistry , Justus Liebig University , Heinrich-Buff-Ring 17 , 35392 Giessen , Germany
| |
Collapse
|
28
|
Wang S, Malebari AM, Greene TF, O'Boyle NM, Fayne D, Nathwani SM, Twamley B, McCabe T, Keely NO, Zisterer DM, Meegan MJ. 3-Vinylazetidin-2-Ones: Synthesis, Antiproliferative and Tubulin Destabilizing Activity in MCF-7 and MDA-MB-231 Breast Cancer Cells. Pharmaceuticals (Basel) 2019; 12:ph12020056. [PMID: 30979033 PMCID: PMC6630832 DOI: 10.3390/ph12020056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/02/2019] [Accepted: 04/07/2019] [Indexed: 12/18/2022] Open
Abstract
Microtubule-targeted drugs are essential chemotherapeutic agents for various types of cancer. A series of 3-vinyl-β-lactams (2-azetidinones) were designed, synthesized and evaluated as potential tubulin polymerization inhibitors, and for their antiproliferative effects in breast cancer cells. These compounds showed potent activity in MCF-7 breast cancer cells with an IC50 value of 8 nM for compound 7s 4-[3-Hydroxy-4-methoxyphenyl]-1-(3,4,5-trimethoxyphenyl)-3-vinylazetidin-2-one) which was comparable to the activity of Combretastatin A-4. Compound 7s had minimal cytotoxicity against both non-tumorigenic HEK-293T cells and murine mammary epithelial cells. The compounds inhibited the polymerisation of tubulin in vitro with an 8.7-fold reduction in tubulin polymerization at 10 μM for compound 7s and were shown to interact at the colchicine-binding site on tubulin, resulting in significant G2/M phase cell cycle arrest. Immunofluorescence staining of MCF-7 cells confirmed that β-lactam 7s is targeting tubulin and resulted in mitotic catastrophe. A docking simulation indicated potential binding conformations for the 3-vinyl-β-lactam 7s in the colchicine domain of tubulin. These compounds are promising candidates for development as antiproiferative microtubule-disrupting agents.
Collapse
Affiliation(s)
- Shu Wang
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Azizah M Malebari
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Thomas F Greene
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Niamh M O'Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Darren Fayne
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Seema M Nathwani
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, 2 DO2R590 Dublin, Ireland.
| | - Thomas McCabe
- School of Chemistry, Trinity College Dublin, 2 DO2R590 Dublin, Ireland.
| | - Niall O Keely
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Mary J Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| |
Collapse
|
29
|
Medicinal chemistry of vicinal diaryl scaffold: A mini review. Eur J Med Chem 2018; 162:1-17. [PMID: 30396033 DOI: 10.1016/j.ejmech.2018.10.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022]
Abstract
The privileged structures have been widely used as a valuable template in new drug discovery. 1,2-Diaryl or vicinal diaryl is a simple scaffold found in many drugs and naturally occurring compounds. From synthetic point of view, the vicinal diaryl derivatives are easily accessible due to their facile and expedient syntheses. These scaffolds have shown numerous interesting pharmacological activities against various diseases with lot of clinical potentials. This review aims to highlight the evidence of vicinal diaryl motif as a privileged scaffold in COX-2 inhibitors and CA-4 analogs.
Collapse
|
30
|
Arun A, Ansari MI, Popli P, Jaiswal S, Mishra AK, Dwivedi A, Hajela K, Konwar R. New piperidine derivative DTPEP acts as dual-acting anti-breast cancer agent by targeting ERα and downregulating PI3K/Akt-PKCα leading to caspase-dependent apoptosis. Cell Prolif 2018; 51:e12501. [PMID: 30091186 DOI: 10.1111/cpr.12501] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 06/02/2018] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES In our ongoing studies to develop ER targeting agents, we screened for dual-acting molecules with a hypothesis that a single molecule can also target both ER positive and negative groups of breast cancer. MATERIALS AND METHODS 1-(2-(4-(Dibenzo[b,f]thiepin-10-yl)phenoxy)ethyl)piperidine (DTPEP) was synthesized and screened in both MCF-7 (ER+ve) and MDA-MB-231 (ER-ve) cells. Assays for analysis of cell cycle, ROS, apoptosis and MMP loss were carried out using flow cytometry. Its target was investigated using western blot, transactivation assay and RT-PCR. In vivo efficacy of DTPEP was validated in LA-7 syngeneic rat mammary tumour model. RESULTS Here, we report identification of dual-acting molecule DTPEP that downregualtes PI3K/Akt and PKCα expression, induces ROS and ROS-dependent apoptosis, loss of mitochondrial membrane potential, induces expression of caspase indicative of both intrinsic and extrinsic apoptosis in MCF-7 and MDA-MB-231 cells. In MCF-7 cells, DTPEP downregulates ERα expression and activation. In MDA-MB-231 cells, primary cellular target of DTPEP is not clearly known, but it downregualtes PI3K/Akt and PKCα expression. In vivo study showed regression of LA-7 syngeneic mammary tumour in SD rat. CONCLUSIONS We identified a new dual-acting anti-breast cancer molecules as a proof of concept which is capable of targeting both ER-positive and ER-negative breast cancer.
Collapse
Affiliation(s)
- A Arun
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - M I Ansari
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - P Popli
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - S Jaiswal
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - A K Mishra
- Department of Endocrine Surgery, King George's Medical University, Lucknow, UP, India
| | - A Dwivedi
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, UP, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Central Drug Research Institute Campus, Lucknow, UP, India
| | - K Hajela
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - R Konwar
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, UP, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Central Drug Research Institute Campus, Lucknow, UP, India
| |
Collapse
|
31
|
Li L, Jiang S, Li X, Liu Y, Su J, Chen J. Recent advances in trimethoxyphenyl (TMP) based tubulin inhibitors targeting the colchicine binding site. Eur J Med Chem 2018; 151:482-494. [PMID: 29649743 DOI: 10.1016/j.ejmech.2018.04.011] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 12/16/2022]
Abstract
Microtubules (composed of α- and β-tubulin heterodimers) play a pivotal role in mitosis and cell division, and are regarded as an excellent target for chemotherapeutic agents to treat cancer. There are four unique binding sites in tubulin to which taxanes, vinca alkaloids, laulimalide and colchicine bind respectively. While several tubulin inhibitors that bind to the taxane or vinca alkaloid binding sites have been approved by FDA, currently there are no FDA approved tubulin inhibitors targeting the colchicine binding site. Tubulin inhibitors that bind to the colchicine binding site have therapeutic advantages over taxanes and vinca alkaloids, for example, they can be administered orally, have less drug-drug interaction potential, and are less prone to develop multi-drug resistance. Typically, tubulin inhibitors that bind to the colchicine binding site bear the trimethoxyphenyl (TMP) moiety which is essential for interaction with tubulin. Over the last decade, a variety of molecules bearing the TMP moiety have been designed and synthesized as tubulin inhibitors for cancer treatment. In this review, we focus on the TMP analogs that are designed based on CA-4, indole, chalcone, colchicine and natural product scaffolds which are known to interact with the colchicine binding site in tubulin. The challenges and future direction of the TMP based tubulin inhibitors are also discussed in detail.
Collapse
Affiliation(s)
- Ling Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Sibo Jiang
- College of Pharmacy, University of Florida, Orlando, FL 32827, USA
| | - Xiaoxun Li
- Chengdu Easton Biopharmaceuticals Co., Ltd., Chengdu 611731, China
| | - Yao Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
32
|
Ibrahim HA, Awadallah FM, Refaat HM, Amin KM. Molecular docking simulation, synthesis and 3D pharmacophore studies of novel 2-substituted-5-nitro-benzimidazole derivatives as anticancer agents targeting VEGFR-2 and c-Met. Bioorg Chem 2018; 77:457-470. [DOI: 10.1016/j.bioorg.2018.01.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 01/12/2023]
|
33
|
Design, synthesis, biological evaluation and cocrystal structures with tubulin of chiral β -lactam bridged combretastatin A-4 analogues as potent antitumor agents. Eur J Med Chem 2018; 144:817-842. [DOI: 10.1016/j.ejmech.2017.12.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/28/2017] [Accepted: 12/02/2017] [Indexed: 11/22/2022]
|
34
|
Zhang M, Liang YR, Li H, Liu MM, Wang Y. Design, synthesis, and biological evaluation of hydantoin bridged analogues of combretastatin A-4 as potential anticancer agents. Bioorg Med Chem 2017; 25:6623-6634. [DOI: 10.1016/j.bmc.2017.10.045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 12/16/2022]
|
35
|
Luo G, Li X, Zhang G, Wu C, Tang Z, Liu L, You Q, Xiang H. Novel SERMs based on 3-aryl-4-aryloxy-2H-chromen-2-one skeleton - A possible way to dual ERα/VEGFR-2 ligands for treatment of breast cancer. Eur J Med Chem 2017; 140:252-273. [DOI: 10.1016/j.ejmech.2017.09.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/24/2017] [Accepted: 09/10/2017] [Indexed: 12/13/2022]
|
36
|
Structure-Activity Relationship Studies of β-Lactam-azide Analogues as Orally Active Antitumor Agents Targeting the Tubulin Colchicine Site. Sci Rep 2017; 7:12788. [PMID: 28986548 PMCID: PMC5630639 DOI: 10.1038/s41598-017-12912-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 09/12/2017] [Indexed: 11/29/2022] Open
Abstract
We have synthesized a series of new β-lactam-azide derivatives as orally active anti-tumor agents by targeting tubulin colchicine binding site and examined their structure activity relationship (SAR). Among them, compound 28 exhibited the most potent antiproliferative activity against MGC-803 cells with an IC50 value of 0.106 μM by induction of G2/M arrest and apoptosis and inhibition of the epithelial to mesenchymal transition. 28 acted as a novel inhibitor of tubulin polymerization by its binding to the colchicine site. SAR analysis revealed that a hydrogen atom at the C-3 position of the β-lactam was required for the potent antiproliferative activity of β-lactam-azide derivatives. Oral administration of compound 28 also effectively inhibited MGC-803 xenograft tumor growth in vivo in nude mice without causing significant loss of body weight. These results suggested that compound 28 is a promising orally active anticancer agent with potential for development of further clinical applications.
Collapse
|
37
|
Malebari AM, Greene LM, Nathwani SM, Fayne D, O'Boyle NM, Wang S, Twamley B, Zisterer DM, Meegan MJ. β-Lactam analogues of combretastatin A-4 prevent metabolic inactivation by glucuronidation in chemoresistant HT-29 colon cancer cells. Eur J Med Chem 2017; 130:261-285. [PMID: 28254699 DOI: 10.1016/j.ejmech.2017.02.049] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/18/2017] [Accepted: 02/20/2017] [Indexed: 11/17/2022]
Abstract
Glucuronidation by uridine 5-diphosphoglucuronosyl transferase enzymes (UGTs) is a cause of intrinsic drug resistance in cancer cells. Glucuronidation of combretastatin A-4 (CA-4) was previously identified as a mechanism of resistance in hepatocellular cancer cells. Herein, we propose chemical manipulation of β-lactam bridged analogues of Combretastatin A-4 as a novel means of overcoming drug resistance associated with glucuronidation due to the expression of UGTs in the CA-4 resistant human colon cancer HT-29 cells. The alkene bridge of CA-4 is replaced with a β-lactam ring to circumvent potential isomerisation while the potential sites of glucuronate conjugation are deleted in the novel 3-substituted-1,4-diaryl-2-azetidinone analogues of CA-4. We hypothesise that glucuronidation of CA-4 is the mechanism of drug resistance in HT-29 cells. Ring B thioether containing 2-azetidinone analogues of CA-4 such as 4-(4-(methylthio)phenyl)-3-phenyl-1-(3,4,5-trimethoxyphenyl)azetidin-2-one (27) and 3-hydroxy-4-(4-(methylthio)phenyl)-1-(3,4,5-trimethoxyphenyl)azetidin-2-one (45) were identified as the most potent inhibitors of tumour cell growth, independent of UGT status, displaying antiproliferative activity in the low nanomolar range. These compounds also disrupted the microtubular structure in MCF-7 and HT-29 cells, and caused G2/M arrest and apoptosis. Taken together, these findings highlight the potential of chemical manipulation as a means of overcoming glucuronidation attributed drug resistance in CA-4 resistant human colon cancer HT-29 cells, allowing the development of therapeutically superior analogues.
Collapse
Affiliation(s)
- Azizah M Malebari
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Dublin 2, Ireland.
| | - Lisa M Greene
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Dublin 2, Ireland
| | - Seema M Nathwani
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Dublin 2, Ireland
| | - Darren Fayne
- Molecular Design Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Niamh M O'Boyle
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Dublin 2, Ireland
| | - Shu Wang
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Dublin 2, Ireland
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Dublin 2, Ireland
| | - Mary J Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
38
|
Olazarán FE, García-Pérez CA, Bandyopadhyay D, Balderas-Rentería I, Reyes-Figueroa AD, Henschke L, Rivera G. Theoretical and experimental study of polycyclic aromatic compounds as β-tubulin inhibitors. J Mol Model 2017; 23:85. [PMID: 28214932 DOI: 10.1007/s00894-017-3256-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/23/2017] [Indexed: 12/14/2022]
Abstract
In this work, through a docking analysis of compounds from the ZINC chemical library on human β-tubulin using high performance computer cluster, we report new polycyclic aromatic compounds that bind with high energy on the colchicine binding site of β-tubulin, suggesting three new key amino acids. However, molecular dynamic analysis showed low stability in the interaction between ligand and receptor. Results were confirmed experimentally in in vitro and in vivo models that suggest that molecular dynamics simulation is the best option to find new potential β-tubulin inhibitors. Graphical abstract Bennett's acceptance ratio (BAR) method.
Collapse
Affiliation(s)
- Fabian E Olazarán
- Facultad de Ciencias Químicas. Av. Universidad s/n, Ciudad Universitaria, San Nicolás de los Garza, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, 64451, México
| | - Carlos A García-Pérez
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Boulevard del Maestro, s/n, Esq. Elías Piña, Reynosa, Tamualipas, Mexico, 88710
| | - Debasish Bandyopadhyay
- Department of Chemistry, The University of Texas Rio Grande Valley, 1201 West University Drive, Edinburg, TX, 78539, USA
| | - Isaias Balderas-Rentería
- Facultad de Ciencias Químicas. Av. Universidad s/n, Ciudad Universitaria, San Nicolás de los Garza, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, 64451, México
| | - Angel D Reyes-Figueroa
- Centro de Investigación y de Estudios Avanzados del Instituto Politecnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, 66600, México
| | - Lars Henschke
- Department of Biology, University of Konstanz, Universitätsstraβe 10, 78457, Konstanz, Germany
| | - Gildardo Rivera
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Boulevard del Maestro, s/n, Esq. Elías Piña, Reynosa, Tamualipas, Mexico, 88710.
| |
Collapse
|
39
|
Banerjee S, Hwang DJ, Li W, Miller DD. Current Advances of Tubulin Inhibitors in Nanoparticle Drug Delivery and Vascular Disruption/Angiogenesis. Molecules 2016; 21:molecules21111468. [PMID: 27827858 PMCID: PMC6272853 DOI: 10.3390/molecules21111468] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/12/2016] [Accepted: 10/27/2016] [Indexed: 01/05/2023] Open
Abstract
Extensive research over the last decade has resulted in a number of highly potent tubulin polymerization inhibitors acting either as microtubule stabilizing agents (MSAs) or microtubule destabilizing agents (MDAs). These inhibitors have potent cytotoxicity against a broad spectrum of human tumor cell lines. In addition to cytotoxicity, a number of these tubulin inhibitors have exhibited abilities to inhibit formation of new blood vessels as well as disrupt existing blood vessels. Tubulin inhibitors as a vascular disrupting agents (VDAs), mainly from the MDA family, induce rapid tumor vessel occlusion and massive tumor necrosis. Thus, tubulin inhibitors have become increasingly popular in the field of tumor vasculature. However, their pharmaceutical application is halted by a number of limitations including poor solubility and toxicity. Thus, recently, there has been considerable interests in the nanoparticle drug delivery of tubulin inhibitors to circumvent those limitations. This article reviews recent advances in nanoparticle based drug delivery for tubulin inhibitors as well as their tumor vasculature disruption properties.
Collapse
Affiliation(s)
- Souvik Banerjee
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave. Memphis, TN 38163, USA.
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave. Memphis, TN 38163, USA.
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave. Memphis, TN 38163, USA.
| | - Duane D Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave. Memphis, TN 38163, USA.
| |
Collapse
|
40
|
Tang Z, Wu C, Wang T, Lao K, Wang Y, Liu L, Muyaba M, Xu P, He C, Luo G, Qian Z, Niu S, Wang L, Wang Y, Xiao H, You Q, Xiang H. Design, synthesis and evaluation of 6-aryl-indenoisoquinolone derivatives dual targeting ERα and VEGFR-2 as anti-breast cancer agents. Eur J Med Chem 2016; 118:328-39. [DOI: 10.1016/j.ejmech.2016.04.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 04/10/2016] [Accepted: 04/11/2016] [Indexed: 11/28/2022]
|
41
|
Carr M, Knox AJS, Lloyd DG, Zisterer DM, Meegan MJ. Development of the β-lactam type molecular scaffold for selective estrogen receptor α modulator action: synthesis and cytotoxic effects in MCF-7 breast cancer cells. J Enzyme Inhib Med Chem 2016; 31:117-130. [PMID: 27476825 DOI: 10.1080/14756366.2016.1210136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The estrogen receptors (ERα and ERβ) which are ligand inducible nuclear receptors are recognized as pharmaceutical targets for diseases such as osteoporosis and breast cancer. There is an increasing interest in the discovery of subtype Selective Estrogen Receptor Modulators (SERMs). A series of novel β-lactam compounds with estrogen receptor modulator properties have been synthesized. The antiproliferative effects of these compounds on human MCF-7 breast tumor cells are reported, together with binding affinity for the ERα and ERβ receptors. The most potent compound 15g demonstrated antiproliferative effects on MCF-7 breast tumor cells (IC50 = 186 nM) and ERα binding (IC50 = 4.3 nM) with 75-fold ERα/β receptor binding selectivity. The effect of positioning of the characteristic amine containing substituted aryl ring (on C-4 or N-1 of the β-lactam scaffold) on the antiproliferative activity and ER-binding properties of the β-lactam compounds is rationalized in a molecular modeling study.
Collapse
Affiliation(s)
- Miriam Carr
- a School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute , Trinity College Dublin , Dublin , Ireland and
| | - Andrew J S Knox
- a School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute , Trinity College Dublin , Dublin , Ireland and.,b School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute , Trinity College Dublin , Dublin , Ireland
| | - David G Lloyd
- b School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute , Trinity College Dublin , Dublin , Ireland
| | - Daniela M Zisterer
- b School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute , Trinity College Dublin , Dublin , Ireland
| | - Mary J Meegan
- a School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute , Trinity College Dublin , Dublin , Ireland and
| |
Collapse
|
42
|
Keely NO, Carr M, Yassin B, Ana G, Lloyd DG, Zisterer D, Meegan MJ. Design, Synthesis and Biochemical Evaluation of Novel Selective Estrogen Receptor Ligand Conjugates Incorporating an Endoxifen-Combretastatin Hybrid Scaffold. Biomedicines 2016; 4:biomedicines4030015. [PMID: 28536383 PMCID: PMC5344255 DOI: 10.3390/biomedicines4030015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/21/2016] [Accepted: 07/07/2016] [Indexed: 02/01/2023] Open
Abstract
Nuclear-receptors are often overexpressed in tumours and can thereby be used as targets when designing novel selective chemotherapeutic agents. To date, many conjugates incorporating an estrogen receptor (ER) ligand have been synthesised in order to direct chemical agents to tissue sites containing ERs. A series of ER ligand conjugates were synthesised incorporating an antagonistic ER ligand scaffold based on endoxifen, covalently-bound via an amide linkage to a variety of combretastatin-based analogues, which may act as antimitotic agents. These novel endoxifen-combretastatin hybrid scaffold analogues were biochemically evaluated in order to determine their antiproliferative and cytotoxicity effects in both the ER-positive MCF-7 and the ER-negative MDA-MB-231 human breast cancer cell lines. ER competitive binding assays were carried out to assess the binding affinity of the lead conjugate 28 towards both the ERα and ERβ isoforms. In results from the NCI 60-cell line screen, the lead conjugate 28 displayed potent and highly selective antiproliferative activity towards the MCF-7 human cancer cell line (IC50 = 5 nM). In the ER-binding assays, the lead conjugate 28 demonstrated potent ER competitive binding in ERα (IC50 value: 0.9 nM) and ERβ (IC50 value: 4.7 nM). Preliminary biochemical results also demonstrate that the lead conjugate 28 may exhibit pure antagonism. This series makes an important addition to the class of ER antagonists and may have potential applications in anticancer therapy.
Collapse
Affiliation(s)
- Niall O Keely
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland.
| | - Miriam Carr
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland.
| | - Bassem Yassin
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland.
| | - Gloria Ana
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Dublin 2, Ireland.
| | - David G Lloyd
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Dublin 2, Ireland.
- Division of Health Sciences, University of South Australia, Adelaide SA 5000, Australia.
| | - Daniela Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Trinity College Dublin, Dublin 2, Ireland.
| | - Mary J Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
43
|
Li C, Tang C, Hu Z, Zhao C, Li C, Zhang S, Dong C, Zhou HB, Huang J. Synthesis and structure–activity relationships of novel hybrid ferrocenyl compounds based on a bicyclic core skeleton for breast cancer therapy. Bioorg Med Chem 2016; 24:3062-3074. [DOI: 10.1016/j.bmc.2016.05.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/12/2016] [Accepted: 05/12/2016] [Indexed: 10/21/2022]
|
44
|
Ameri A, Khodarahmi G, Hassanzadeh F, Forootanfar H, Hakimelahi GH. Novel Aldimine-Type Schiff Bases of 4-Amino-5-[(3,4,5-trimethoxyphenyl)methyl]-1,2,4-triazole-3-thione/thiol: Docking Study, Synthesis, Biological Evaluation, and Anti-Tubulin Activity. Arch Pharm (Weinheim) 2016; 349:662-81. [DOI: 10.1002/ardp.201600021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 05/07/2016] [Accepted: 05/20/2016] [Indexed: 01/15/2023]
Affiliation(s)
- Alieh Ameri
- Department of Medicinal Chemistry; Faculty of Pharmacy; Isfahan University of Medical Sciences; Isfahan Iran
| | - Ghadamali Khodarahmi
- Department of Medicinal Chemistry; Faculty of Pharmacy; Isfahan University of Medical Sciences; Isfahan Iran
| | - Farshid Hassanzadeh
- Department of Medicinal Chemistry; Faculty of Pharmacy; Isfahan University of Medical Sciences; Isfahan Iran
| | - Hamid Forootanfar
- Department of Pharmaceutical Biotechnology; Faculty of Pharmacy; Kerman University of Medical Sciences; Kerman Iran
| | - Gholam-Hosein Hakimelahi
- Department of Medicinal Chemistry; Faculty of Pharmacy; Isfahan University of Medical Sciences; Isfahan Iran
- Institute of Chemistry; Academia Sinica; Nankang Taipei Taiwan
| |
Collapse
|
45
|
|