1
|
Fotopoulos I, Hadjipavlou-Litina D. Approaches for the discovery of cinnamic acid derivatives with anticancer potential. Expert Opin Drug Discov 2024; 19:1281-1291. [PMID: 39105559 DOI: 10.1080/17460441.2024.2387122] [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: 05/20/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
INTRODUCTION Cinnamic acid is a privileged scaffold for the design of biologically active compounds with putative anticancer potential, following different synthetic methodologies and procedures. Since there is a need for the production of potent anticancer, cinnamate moiety can significantly contribute in the design of new and more active anticancer agents. AREAS COVERED In this review, the authors provide a review on the synthetic approaches for the discovery of cinnamic acid derivatives with anticancer potential. Results from molecular simulations, hybridization, and chemical derivatization along with biological experiments in vitro and structural activity relationships are given, described, and discussed by the authors. Information for the mechanism of action is taken from original literature sources. EXPERT OPINION The authors suggest that (i) numerous areas of biology-pharmacology need to be considered: selectivity, in vivo studies, toxicity and drug-likeness, the mechanism of action in animals and humans, development of more efficient assays for various cancer types; (ii) hybridization techniques outbalance in the discovery and production of compounds with higher activity and greater selectivity; (iii) repositioning offers new anticancer cinnamic agents.
Collapse
Affiliation(s)
- Ioannis Fotopoulos
- Department of Pharmaceutical Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | |
Collapse
|
2
|
Fu XJ, Huang J, Li N, Liu YH, Liu QG, Yuan S, Xu Y, Chen YF, Zhao YX, Song J, Zhang SY, Bai YR. Design, synthesis and biological evaluation of N-benzylaryl cinnamide derivatives as tubulin polymerization inhibitors capable of promoting YAP degradation with potent anti-gastric cancer activities. Eur J Med Chem 2023; 262:115883. [PMID: 39491429 DOI: 10.1016/j.ejmech.2023.115883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/07/2023] [Accepted: 10/14/2023] [Indexed: 11/05/2024]
Abstract
In this work, we utilized the N-benzylaryl derivative 9 as a lead compound and employed the molecular hybridization strategy by introducing cinnamoyl fragments to successfully design and synthesize 33 novel N-benzylaryl cinnamide derivatives 15a∼15 ag. The in vitro antiproliferative activities were explored, and the preliminary analysis and summary of their structure-activity relationship were conducted. The majority of the compounds demonstrated significant inhibitory potency on MGC-803, HCT-116 and KYSE450 cells with IC50 values below 0.5 μM. Among them, compound 15e (MY-1076) exhibited the most effective effect on the proliferative inhibition of MGC-803, SGC-7901, HCT-116 and KYSE450 cells with IC50 values of 0.019, 0.017, 0.020 and 0.044 μM, respectively, which is more potent than colchicine and the lead compound 9. Additionally, compound 15e (MY-1076) still exhibited significant inhibitory proliferation activity against 13 other types of tumor cells (IC50 values < 0.1 μM). Further studies revealed that compound 15e (MY-1076) could effectively inhibit tubulin polymerization by acting on the β-tubulin colchicine binding site, thereby disrupting microtubule network assembly and mitotic progression. Additionally, compound 15e (MY-1076) also demonstrated a notable inhibitory effect on the oncogenic protein YAP by inducing its degradation. Compound 15e (MY-1076) could dose-dependently induce G2/M phase arrest and cell apoptosis, effectively inhibit the colony formatting ability and cause morphological changes in MGC-803 and SGC-7901 cells. Compound 15e (MY-1076) exhibited significant regulatory effects on the expression levels of cell cycle and apoptosis-related proteins. Taken together, we here reported a novel N-benzylaryl cinnamide derivative 15e (MY-1076) as a tubulin polymerization inhibitor capable of promoting degradation of YAP, which held great potential as an anti-gastric cancer agent.
Collapse
Affiliation(s)
- Xiang-Jing Fu
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China; School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Jiao Huang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Na Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Yun-He Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Qiu-Ge Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Shuo Yuan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China; School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Yan Xu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yi-Fan Chen
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yu-Xuan Zhao
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yi-Ru Bai
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China.
| |
Collapse
|
3
|
Villamizar–Mogotocoro A, Kouznetsov VV. Simple and efficient microwave-assisted synthesis of new N-biphenyl cinnamamides/3-arylpropanamides and C6-substituted phenanthridines. Tetrahedron Lett 2023. [DOI: 10.1016/j.tetlet.2023.154461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
4
|
González M, Ellahioui Y, Gallego L, Vicente-Blázquez A, Álvarez R, Medarde M, Peláez R. Novel amino analogs of the trimethoxyphenyl ring in potent colchicine site ligands improve solubility by the masked polar group incorporation (MPGI) strategy. Bioorg Chem 2023; 131:106282. [PMID: 36459777 DOI: 10.1016/j.bioorg.2022.106282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
Abstract
The low aqueous solubility of colchicine site antimitotic agents, of which the trimethoxyphenyl (A ring) is a heavy contributor, is a serious drawback in their clinical development. We have designed new A ring analogs with chameleonic masked polar amino groups able to increase aqueous solubility and also behave as non-polar through intramolecular hydrogen bonds when bound to tubulin. We have incorporated these new A rings in several scaffolds (sulfonamides, combretastatins, phenstatins, isocombretastatins), synthesized, and assayed 43 representatives. The amino analogs show improved aqueous solubility and some of them (8, 60Z, and 67) nanomolar anti-proliferative potencies against human cancer cell lines, with the most favorable substituent being a 3-methylamino group. The antiproliferative effect relates to tubulin inhibition as shown by in vitro tubulin polymerization inhibition, immunofluorescence microscopy, and cell cycle and apoptosis analysis by flow cytometry. The compounds arrest the cell cycle of treated cells in G2/M and later develop an apoptotic response. Docking studies suggested binding at the colchicine site of tubulin with good agreement with the DFT models of the new structural variations made. The 3-methylamino-4,5‑dimethoxyphenyl moiety is an example of the masked polar group incorporation (MPGI) strategy for soluble ligands binding to hydrophobic sites and a good trimethoxyphenyl ring replacement for the development of new colchicine site ligands.
Collapse
Affiliation(s)
- Myriam González
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Younes Ellahioui
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Laura Gallego
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Alba Vicente-Blázquez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Manuel Medarde
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain.
| |
Collapse
|
5
|
Mirzaei S, Ghodsi R, Hadizadeh F, Sahebkar A. 3D-QSAR-Based Pharmacophore Modeling, Virtual Screening, and Molecular Docking Studies for Identification of Tubulin Inhibitors with Potential Anticancer Activity. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6480804. [PMID: 34485522 PMCID: PMC8410400 DOI: 10.1155/2021/6480804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/22/2021] [Indexed: 11/29/2022]
Abstract
In this study, we aimed to develop a pharmacophore-based three-dimensional quantitative structure activity relationship (3D-QSAR) for a set including sixty-two cytotoxic quinolines (1-62) as anticancer agents with tubulin inhibitory activity. A total of 279 pharmacophore hypotheses were generated based on the survival score to build QSAR models. A six-point pharmacophore model (AAARRR.1061) was identified as the best model which consisted of three hydrogen bond acceptors (A) and three aromatic ring (R) features. The model showed a high correlation coefficient (R 2 = 0.865), cross-validation coefficient (Q 2 = 0.718), and F value (72.3). The best pharmacophore model was then validated by the Y-Randomization test and ROC-AUC analysis. The generated 3D contour maps were used to reveal the structure activity relationship of the compounds. The IBScreen database was screened against AAARRR.1061, and after calculating ADMET properties, 10 compounds were selected for further docking study. Molecular docking analysis showed that compound STOCK2S-23597 with the highest docking score (-10.948 kcal/mol) had hydrophobic interactions and can form four hydrogen bonds with active site residues.
Collapse
Affiliation(s)
- Salimeh Mirzaei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Razieh Ghodsi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Tang L, Chen T, Yang H, Wen X, Sun Y, Liu S, Peng T, Zhang S, Wang L. Synthesis and antitumor effects of novel benzyl naphthyl sulfoxide/sulfone derivatives derived from Rigosertib. RSC Adv 2021; 11:37462-37471. [PMID: 35496445 PMCID: PMC9043816 DOI: 10.1039/d1ra05226h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/14/2021] [Indexed: 11/21/2022] Open
Abstract
In this work, a series of novel benzyl naphthyl sulfoxides/sulfones derived from Rigosertib were designed and synthesized as potential antitumor agents. The in vitro cytotoxicity against four human cancer cell lines (HeLa, MCF-7, HepG2 and SCC-15) and two normal human cell lines (HUVEC and 293T) indicated that some of the sulfones and sulfoxides possessed potent antineoplastic activity that reached nanomolar levels and relatively low toxicity to normal cells. Among them, (2-methoxy-5-((naphthalen-2-ylsulfonyl)methyl)phenyl)glycine (15b) was found to be a promising antitumor drug candidate that could significantly inhibit tumor cell migration and induce tumor cell apoptosis via the p53-Bcl-2-Bax signaling pathway at nanomolar concentrations. In this work, a series of novel benzyl naphthyl sulfoxides/sulfones derived from Rigosertib were designed and synthesized as potential antitumor agents.![]()
Collapse
Affiliation(s)
- Lin Tang
- Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Tingting Chen
- Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Hongpeng Yang
- Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
- Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, P. R. China
| | - Xiaoxue Wen
- Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Yunbo Sun
- Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Shuchen Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Tao Peng
- Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Shouguo Zhang
- Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Lin Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
- Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, P. R. China
| |
Collapse
|
7
|
Sethi NS, Prasad DN, Singh RK. An Insight into the Synthesis and SAR of 2,4-Thiazolidinediones (2,4-TZD) as Multifunctional Scaffold: A Review. Mini Rev Med Chem 2020; 20:308-330. [PMID: 31660809 DOI: 10.2174/1389557519666191029102838] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 10/11/2019] [Accepted: 10/19/2019] [Indexed: 01/18/2023]
Abstract
2,4-Thiazolidinedione (2,4-TZD) is a versatile pharmacophore, a privileged scaffold, and a remarkable sulphur-containing heterocyclic compound with diverse pharmacological activities. The multifarious biological activities, due to different mechanisms of action, low cost, and easy availability of 2,4-TZD impressed medicinal chemists to integrate this moiety to develop various lead compounds with diverse therapeutic actions. This resulted in the swift development in the last decade for generating different new potential molecules bearing 2,4-TZD. In this review, the authors attempt to shape and present the latest investigations (2012 onwards) going on in generating promising 2,4-TZD containing lead compounds. The data has been collected and analyzed to develop the structure-activity relationship (SAR). The SAR and active pharmacophores of various leads accountable for antidiabetic, anticancer, antimicrobial, and antioxidant activities have also been illustrated. This review also highlighted some of the important chemical synthetic routes for the preparation of various 2,4-TZD derivatives. This review will definitely serve as a useful source of structural information to medicinal chemists and may be utilized for the strategic design of potent 2,4-TZD derivatives in the future.
Collapse
Affiliation(s)
- Navjot Singh Sethi
- School of Pharmacy, Maharaja Agrasen University, Baddi, Dist Solan, 160022, Himachal Pradesh, India.,Faculty of Pharmacy, I.K. Gujral Punjab Technical University, Jalandhar, India
| | - Deo Nandan Prasad
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Dist. Rupnagar, 140126, Punjab, India
| | - Rajesh Kumar Singh
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Dist. Rupnagar, 140126, Punjab, India
| |
Collapse
|
8
|
Khan MF, Verma G, Akhtar W, Shaquiquzzaman M, Akhter M, Rizvi MA, Alam MM. Pharmacophore modeling, 3D-QSAR, docking study and ADME prediction of acyl 1,3,4-thiadiazole amides and sulfonamides as antitubulin agents. ARAB J CHEM 2019. [DOI: 10.1016/j.arabjc.2016.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
9
|
Gaikwad N, Nanduri S, Madhavi YV. Cinnamamide: An insight into the pharmacological advances and structure-activity relationships. Eur J Med Chem 2019; 181:111561. [PMID: 31376564 DOI: 10.1016/j.ejmech.2019.07.064] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/11/2019] [Accepted: 07/23/2019] [Indexed: 01/07/2023]
Abstract
The cinnamamide (cinnamic acid amide and cinnamide) is a privileged scaffold present widely in a number of natural products. The scaffold acts as a useful template for designing and arriving at newly drug-like molecules with potential pharmacological activity. An attempt has been made to review the extensive occurrence of cinnamamide scaffold in many lead compounds reported for treating various diseases, their binding interactions with the therapeutic targets as well as mechanism of action and their structure-activity relationships. The discoveries of cinnamamide systems and some examples of unusual cinnamamides having an aromatic, aliphatic, and heterocyclic or other rings condensed to the basic cinnamamide structure also have been extensively covered in this review.
Collapse
Affiliation(s)
- Nikhil Gaikwad
- Department of Pharmaceutical Technology and Process Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), 40, Dilip Rd, Jinkalwada, Balanagar, Hyderabad, Telangana, 500037, India
| | - Srinivas Nanduri
- Department of Pharmaceutical Technology and Process Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), 40, Dilip Rd, Jinkalwada, Balanagar, Hyderabad, Telangana, 500037, India
| | - Y V Madhavi
- Department of Pharmaceutical Technology and Process Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), 40, Dilip Rd, Jinkalwada, Balanagar, Hyderabad, Telangana, 500037, India.
| |
Collapse
|
10
|
Hybrid cis-stilbene Molecules: Novel Anticancer Agents. Int J Mol Sci 2019; 20:ijms20061300. [PMID: 30875859 PMCID: PMC6471163 DOI: 10.3390/ijms20061300] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/10/2019] [Accepted: 03/11/2019] [Indexed: 12/15/2022] Open
Abstract
The growing interest in anticancer hybrids in the last few years has resulted in a great number of reports on hybrid design, synthesis and bioevaluation. Many novel multi-target-directed drug candidates were synthesized, and their biological activities were evaluated. For the design of anticancer hybrid compounds, the molecules of stilbenes, aromatic quinones, and heterocycles (benzimidazole, imidazole, pyrimidine, pyridine, pyrazole, quinoline, quinazoline) were applied. A distinct group of hybrids comprises the molecules built with natural compounds: Resveratrol, curcumin, coumarin, and oleanolic acid. In this review, we present the studies on bioactive hybrid molecules of a well-known tubulin polymerization inhibitor, combretastatin A-4 and its analogs with other pharmacologically active entities. The mechanism of anticancer activity of selected hybrids is discussed considering the structure-activity relationship.
Collapse
|
11
|
Pang Y, Yan J, An B, Huang L, Li X. The synthesis and evaluation of new butadiene derivatives as tubulin polymerization inhibitors. Bioorg Med Chem 2017; 25:3059-3067. [DOI: 10.1016/j.bmc.2017.03.066] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/24/2017] [Accepted: 03/26/2017] [Indexed: 10/19/2022]
|
12
|
Zghaib Z, Guichou JF, Vappiani J, Bec N, Hadj-Kaddour K, Vincent LA, Paniagua-Gayraud S, Larroque C, Moarbess G, Cuq P, Kassab I, Deleuze-Masquéfa C, Diab-Assaf M, Bonnet PA. New imidazoquinoxaline derivatives: Synthesis, biological evaluation on melanoma, effect on tubulin polymerization and structure-activity relationships. Bioorg Med Chem 2016; 24:2433-2440. [PMID: 27094151 DOI: 10.1016/j.bmc.2016.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/25/2016] [Accepted: 04/01/2016] [Indexed: 12/14/2022]
Abstract
Microtubules are considered as important targets of anticancer therapy. EAPB0503 and its structural imidazo[1,2-a]quinoxaline derivatives are major microtubule-interfering agents with potent anticancer activity. In this study, the synthesis of several new derivatives of EAPB0503 is described, and the anticancer efficacy of 13 novel derivatives on A375 human melanoma cell line is reported. All new compounds show significant antiproliferative activity with IC50 in the range of 0.077-122μM against human melanoma cell line (A375). Direct inhibition of tubulin polymerization assay in vitro is also assessed. Results show that compounds 6b, 6e, 6g, and EAPB0503 highly inhibit tubulin polymerization with percentages of inhibition of 99%, 98%, 90%, and 84% respectively. Structure-activity relationship studies within the series are also discussed in line with molecular docking studies into the colchicine-binding site of tubulin.
Collapse
Affiliation(s)
- Zahraa Zghaib
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, Faculté de Pharmacie, 15, avenue Charles Flahault, BP14491, 34093 Montpellier cedex 5, France; Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, BP 90656, Fanar Jdeideh, Lebanon
| | - Jean-François Guichou
- CNRS, UMR5048 - Université de Montpellier, Centre de Biochimie Structurale, F-34090 Montpellier, France; INSERM, U1054, F-34090 Montpellier, France
| | - Johanna Vappiani
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, Faculté de Pharmacie, 15, avenue Charles Flahault, BP14491, 34093 Montpellier cedex 5, France
| | - Nicole Bec
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U896, 34298 Montpellier, France
| | - Kamel Hadj-Kaddour
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, Faculté de Pharmacie, 15, avenue Charles Flahault, BP14491, 34093 Montpellier cedex 5, France
| | - Laure-Anaïs Vincent
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, Faculté de Pharmacie, 15, avenue Charles Flahault, BP14491, 34093 Montpellier cedex 5, France
| | - Stéphanie Paniagua-Gayraud
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, Faculté de Pharmacie, 15, avenue Charles Flahault, BP14491, 34093 Montpellier cedex 5, France
| | - Christian Larroque
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U896, 34298 Montpellier, France
| | - Georges Moarbess
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, BP 90656, Fanar Jdeideh, Lebanon
| | - Pierre Cuq
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, Faculté de Pharmacie, 15, avenue Charles Flahault, BP14491, 34093 Montpellier cedex 5, France
| | - Issam Kassab
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, BP 90656, Fanar Jdeideh, Lebanon
| | - Carine Deleuze-Masquéfa
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, Faculté de Pharmacie, 15, avenue Charles Flahault, BP14491, 34093 Montpellier cedex 5, France.
| | - Mona Diab-Assaf
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, BP 90656, Fanar Jdeideh, Lebanon
| | - Pierre-Antoine Bonnet
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, Faculté de Pharmacie, 15, avenue Charles Flahault, BP14491, 34093 Montpellier cedex 5, France
| |
Collapse
|
13
|
Zabiulla, Shamanth Neralagundi HG, Bushra Begum A, Prabhakar BT, Khanum SA. Design and synthesis of diamide-coupled benzophenones as potential anticancer agents. Eur J Med Chem 2016; 115:342-51. [PMID: 27027818 DOI: 10.1016/j.ejmech.2016.03.040] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 12/19/2022]
Abstract
A series of diamide-coupled benzophenone, 2-(4-benzoyl-phenoxy)-N-{2-[2-(4-benzoyl-phenoxy)-acetylamino]-phenyl}-acetamide analogues (9a-l) were synthesized by multistep reactions and all compounds were well characterized. Among the series (9a-l), compound 9k with three methyl groups at ortho position in rings A, B, and D and bromo group at the para position in ring E was selected as a lead compound by screening through multiple cancer cell types by in-vitro cytotoxic and antiproliferative assay systems. Also, the cytotoxic nature of the compound 9k resulted the regression of the tumor growth in-vivo, which could be due to decreased vascularisation in the peritoneum lining of the mice which regress the tumor growth. The results were reconfirmed in-vivo chorioallantoic membrane model which indicates a scope of developing 9k into potent anticancer drug in near future.
Collapse
Affiliation(s)
- Zabiulla
- Department of Chemistry, Yuvaraja's College (Autonomous), University of Mysore, Mysore, Karnataka, India
| | - H G Shamanth Neralagundi
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Karnataka, India
| | - A Bushra Begum
- Department of Chemistry, Yuvaraja's College (Autonomous), University of Mysore, Mysore, Karnataka, India
| | - B T Prabhakar
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Karnataka, India
| | - Shaukath Ara Khanum
- Department of Chemistry, Yuvaraja's College (Autonomous), University of Mysore, Mysore, Karnataka, India.
| |
Collapse
|
14
|
Kamal A, Bajee S, Lakshma Nayak V, Venkata Subba Rao A, Nagaraju B, Ratna Reddy C, Jeevak Sopanrao K, Alarifi A. Synthesis and biological evaluation of arylcinnamide linked combretastatin-A4 hybrids as tubulin polymerization inhibitors and apoptosis inducing agents. Bioorg Med Chem Lett 2016; 26:2957-2964. [PMID: 27161282 DOI: 10.1016/j.bmcl.2016.03.049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/26/2016] [Accepted: 03/14/2016] [Indexed: 10/22/2022]
Abstract
A series of new molecules have been designed based on a hybridization approach by combining the arylcinnamide and combretastatin pharmacophores. These were synthesized and evaluated for their cytotoxic activity, effect on inhibition of tubulin polymerization and apoptosis inducing ability. Most of the conjugates exhibited significant cytotoxic activity against some representative human cancer cell lines and two of the conjugates 6i and 6p displayed potent cytotoxicity with GI50 values of 56nM and 31nM respectively against the human breast cancer cell line (MCF-7). SAR studies revealed that 3,4-substitution on the phenyl ring of the cinnamide moiety is beneficial for enhanced cytotoxicity. Moreover, G2/M cell cycle arrest was induced by these conjugates (6i and 6p) apart from tubulin polymerization inhibition (IC50 of 1.97μM and 1.05μM respectively). Further, mitochondrial membrane potential, Annexin V-FITC and caspase-9 activation assays suggested that these conjugates induce cell death by apoptosis. Docking studies revealed that these conjugates interact and bind at the colchicine binding site of the tubulin.
Collapse
Affiliation(s)
- Ahmed Kamal
- Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India; Chemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Shaik Bajee
- Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Vadithe Lakshma Nayak
- Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | | | - Burri Nagaraju
- Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Challa Ratna Reddy
- Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Kapure Jeevak Sopanrao
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Abdullah Alarifi
- Chemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
15
|
Recent developments in tubulin polymerization inhibitors: An overview. Eur J Med Chem 2014; 87:89-124. [DOI: 10.1016/j.ejmech.2014.09.051] [Citation(s) in RCA: 210] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/11/2014] [Accepted: 09/14/2014] [Indexed: 12/11/2022]
|
16
|
P. Brown D, Zhao H, M. Khondoker J, R. Bhavsar J, C. Sigamoney C. Synthesis of Novel β-Lactam Hybrids of Phenstatin and Other Substituted Aromatics as New Bioactives. HETEROCYCLES 2014. [DOI: 10.3987/com-14-13085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|