1
|
Liu YH, Wang ZY, Du YF, Liu XH, Niu JB, Song J, Jin CY, Zhang SY. Thienopyrimidine: A promising scaffold in the development of kinase inhibitors with anticancer activities. Bioorg Med Chem 2025; 121:118109. [PMID: 39955801 DOI: 10.1016/j.bmc.2025.118109] [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: 11/18/2024] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Protein kinases represent a highly promising drug target, with over 80 drugs that target about two dozen different protein kinases have been approved by the US FDA, particularly in cancer treatment. Over the past decades, the unique structural characteristics of the thienopyrimidine ring system provide an adaptive platform for designing potent anticancer agents, especially various kinase inhibitors, which has attracted widespread attention. Some of these thienopyrimidines as anticancer kinase inhibitors have already been marketed or are currently undergoing clinical/preclinical studies for the treatment of cancers, such as Olmutinib, Pictilisib, SNS-314, PF-03758309, and Fimepinostat, highlighting the substantial advantages of the thienopyrimidine scaffold in the discovery of anticancer agents. This article reviews the discovery, activity, and structure-activity relationships of antitumor kinase inhibitors based on the thienopyrimidine scaffold, and partially discusses the binding modes between thienopyrimidine derivatives and their kinase targets. By elucidating the application of thienopyrimidine derivatives as anticancer kinase inhibitors, this review aims to provide new perspectives for the development of more effective and novel kinase inhibitors.
Collapse
Affiliation(s)
- 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
| | - Zi-Yue Wang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China
| | - Yi-Fei Du
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xuan-Han Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jin-Bo Niu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Cheng-Yun Jin
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
2
|
Valipour M, Zakeri Khatir Z, Ayati A, Hosseini A, Sheibani M, Irannejad H. Advances in the selective c-MET kinase inhibitors: Application of fused [5,6]-Bicyclic nitrogen-containing cores for anticancer drug design. Eur J Med Chem 2025; 284:117177. [PMID: 39724725 DOI: 10.1016/j.ejmech.2024.117177] [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: 09/26/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Over the past two decades, small molecules bearing [5,6]-bicyclic nitrogen-containing cores have emerged as one of the most extensively studied structures for the development of selective c-MET kinase inhibitors. Structure-activity relationship (SAR) studies have demonstrated that modifying these cores can significantly impact the biological properties of c-MET inhibitors, including safety/toxicity, potency, and metabolic stability. For example, although c-MET kinase inhibitors containing the [1,2,4]triazolo[4,3-b][1,2,4]triazine scaffold (core P) exhibit high inhibitory potency, they often face challenges due to metabolic stability defects. Alternatively, compounds containing [1,2,3]triazolo[4,5-b]pyrazine (core K) and [1,2,4]triazolo[4,3-b]pyridazine (core I) scaffolds demonstrate lower potency but improved metabolic stability, allowing some of them to progress into clinical trials and even be approved as novel anticancer drugs. Fortunately, X-ray crystallography studies have well elucidated key interactions between [5,6]-bicyclic nitrogen-containing cores and crucial amino acid residues within the c-MET active site. These insights emphasize the significance of π-π stacking interactions with Tyr1230 and hydrogen bonding with Asp1222, providing valuable guidance for the targeted design and optimization of selective c-MET kinase inhibitors. Following the identification/introduction of sixteen distinct [5,6]-bicyclic nitrogen-containing cores (cores A-P) utilized in the design of selective c-MET kinase inhibitors over the past two decades, this manuscript offers a comprehensive review of their roles in drug development of anticancer agents, and describes the various synthesis methods employed. The insights presented herein can serve as a resource for better structural optimization of c-MET kinase inhibitors in the future research.
Collapse
Affiliation(s)
- Mehdi Valipour
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Zahra Zakeri Khatir
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Adileh Ayati
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asieh Hosseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sheibani
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hamid Irannejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
3
|
Mortaheb S, Pezeshki PS, Rezaei N. Bispecific therapeutics: a state-of-the-art review on the combination of immune checkpoint inhibition with costimulatory and non-checkpoint targeted therapy. Expert Opin Biol Ther 2024; 24:1335-1351. [PMID: 39503381 DOI: 10.1080/14712598.2024.2426636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have revolutionized the field of cancer immunotherapy and have enhanced the survival of patients with malignant tumors. However, the overall efficacy of ICIs remains unsatisfactory and is faced with two major concerns of resistance development and occurrence of immune-related adverse events (irAEs). Bispecific antibodies (bsAbs) have emerged as promising strategies with unique mechanisms of action to achieve a better efficacy and safety than monoclonal antibodies (mAbs) or even their combination. BsAbs along with other bispecific platforms such as bispecific fusion proteins, nanobodies, and CAR-T cells may help to avoid development of resistance and reduce irAEs caused by on-target/off-tumor binding effects of mAbs. AREAS COVERED A literature search was performed using PubMed for English-language articles to provide a comprehensive overview of preclinical and clinical studies on bsAbs specified for both immune checkpoints and non-checkpoint molecules as a well-enhanced class of therapeutics. EXPERT OPINION Identifying suitable targets and selecting effective engineering platforms enhance the potential of bsAbs to address the challenges associated with conventional therapies such as ICIs, positioning them as a promising class of therapeutics in the landscape of cancer immunotherapy.
Collapse
Affiliation(s)
- Samin Mortaheb
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parmida Sadat Pezeshki
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
DeAzevedo R, Steiner M, Turner BX, Liu A, Newton S, Schmidt J, Fleming R, Tolentino A, Kaseb AO, Curran MA. Type I MET inhibitors cooperate with PD-1 blockade to promote rejection of hepatocellular carcinoma. J Immunother Cancer 2024; 12:e009690. [PMID: 39477243 PMCID: PMC11529525 DOI: 10.1136/jitc-2024-009690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/04/2024] [Indexed: 11/03/2024] Open
Abstract
Blockade of the immune checkpoints programmed death-1 (PD-1) and cytotoxic lymphocyte antigen 4 has improved outcomes for patients with hepatocellular carcinoma (HCC), yet most still fail to achieve objective clinical benefit. MET plays key roles in both HCC tumorigenesis and immunosuppressive conditioning; however, inhibition of MET causes upregulation of PD-ligand 1 (PD-L1) suggesting the use of these inhibitors in the context of PD-1 blockade. We sought to investigate across the Hepa1-6, HCA-1 and diethylnitrosamine (DEN) models of HCC whether the combination of more specific type I versus more pleiotropic type II MET inhibitors would confer superior outcomes in combination with PD-1 blockade. While MET inhibition demonstrated cooperativity with αPD-1 across all three models, the type I MET inhibitor capmatinib showed optimal activity in combination and statistically significantly outperformed the combination with the type II inhibitor cabozantinib in the αPD-1 refractory DEN model. In both HCA-1 and DEN HCC, the capmatinib and αPD-1 combination enhanced CD8 T cell frequency and activation state while limiting intratumoral myeloid immune suppression. In vitro studies of antigen-specific T cell activation reveal significantly less inhibition of effector cytokine production and proliferation by capmatinib versus by type II or type III MET inhibitors. These findings suggest significant potential for clinical HCC combination studies of type I MET inhibitors and PD-1 blockade where prior trials using type II inhibitors have yielded limited benefit.
Collapse
Affiliation(s)
- Ricardo DeAzevedo
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Madeline Steiner
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Broderick X Turner
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Arthur Liu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Sherwin Newton
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | - Ahmed O Kaseb
- Department of Gastrointestinal (GI) Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael A Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
5
|
Iweala EEJ, Amuji DN, Oluwajembola AM, Ugbogu EA. Targeting c-Met in breast cancer: From mechanisms of chemoresistance to novel therapeutic strategies. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 7:100204. [PMID: 39524211 PMCID: PMC11543557 DOI: 10.1016/j.crphar.2024.100204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/28/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Breast cancer presents a significant challenge due to its heterogeneity and propensity for developing chemoresistance, particularly in the triple-negative subtype. c-Mesenchymal epithelial transition factor (c-Met), a receptor tyrosine kinase, presents a promising target for breast cancer therapy due to its involvement in disease progression and poor prognosis. However, the heterogeneous expression of c-Met within breast cancer subtypes and individual tumors complicates targeted therapy. Also, cancer cells can develop resistance to c-Met inhibitors through various mechanisms, including bypass signaling pathways and genetic mutations. The off-target effects of c-Met inhibitors further limit their clinical utility, necessitating the development of more selective agents. To overcome these challenges, personalized treatment approaches and combination therapies are being explored to improve treatment efficacy while minimizing adverse effects. Novel c-Met inhibitors with improved selectivity and reduced off-target toxicity show promise in preclinical studies. Additionally, targeted delivery systems aim to enhance drug localization and reduce systemic toxicity. Future directions involve refining inhibitor design and integrating c-Met inhibition into personalized treatment regimens guided by molecular profiling. This review explores the mechanisms by which c-Met contributes to chemoresistance in breast cancer and current challenges in targeting c-Met for breast cancer therapy. It discusses strategies to optimize treatment outcomes, ultimately improving patient prognosis and reducing mortality rates associated with this devastating disease.
Collapse
Affiliation(s)
- Emeka Eze Joshua Iweala
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Doris Nnenna Amuji
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Abimbola Mary Oluwajembola
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | | |
Collapse
|
6
|
Vahabi M, Xu G, Avan A, Peters GJ, Giovannetti E. Pharmacological mechanisms underlying the interaction of the nucleoside analogue gemcitabine with the c-MET inhibitor tivantinib in pancreatic cancer. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 43:837-850. [PMID: 38420938 DOI: 10.1080/15257770.2024.2319215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/06/2024] [Accepted: 02/11/2024] [Indexed: 03/02/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly malignancy with limited treatment options, highlighting the urgent need for innovative approaches. A promising target for new anticancer therapies across various tumor types is the receptor tyrosine kinase c-MET. Here, we examined the impact of the c-MET inhibitor tivantinib in combination with gemcitabine on both primary and immortalized PDAC cells, and we investigated the mechanism underlying this combined treatment's effects. Our findings demonstrate that tivantinib is synergistic with gemcitabine, which is not related to cytidine deaminase but to inhibition of the polymerization of tubulin. Moreover, these drugs affected the expression of microRNAs miR-21 and miR-34, which regulate key oncogenic pathways. These findings might have an impact on the selection of patients for future trials.
Collapse
Affiliation(s)
- Mahrou Vahabi
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Geng Xu
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Godefridus J Peters
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Fondazione Pisana per la Scienza, Pisa, Italy
| |
Collapse
|
7
|
Daoui O, Nour H, Abchir O, Elkhattabi S, Bakhouch M, Chtita S. A computer-aided drug design approach to explore novel type II inhibitors of c-Met receptor tyrosine kinase for cancer therapy: QSAR, molecular docking, ADMET and molecular dynamics simulations. J Biomol Struct Dyn 2023; 41:7768-7785. [PMID: 36120976 DOI: 10.1080/07391102.2022.2124456] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/09/2022] [Indexed: 10/14/2022]
Abstract
Small molecules such as 4-phenoxypyridine derivatives have remarkable inhibitory activity against c-Met enzymatic activity and proliferation of cancer cell lines. Since there is a relationship between structure and biological activity of these molecules, these little compounds may have great potential for clinical pharmaceutical use against various types of cancer caused by c-Met activity. The purpose of this study was to remodel the structures of 4-phenoxypyridine derivatives to achieve strong inhibitory activity against c-Met and provide favorable pharmacokinetic properties for drug design and discovery. Therefore, this paper describes the structure-activity relationship and the rationalization of appropriate pharmacophore sites to improve the biological activity of the investigated molecules, based on bioinformatics techniques represented by a computer-aided drug design approach. Accordingly, robust and reliable 3D-QSAR models were developed based on CoMFA and CoMSIA techniques. As a result, 46 lead molecules were designed and their biological and pharmacokinetic activities were predicted in silico. Screening filters by 3D-QSAR, Molecular Docking, drug-like and ADME-Tox identified the computer-designed compounds P54 and P55 as the best candidates to achieve high inhibition of c-Met enzymatic activity compared to the synthesized template compound T14. Finally, through molecular dynamics simulations, the structural properties and dynamics of c-Met free and complex (PDB code: 3LQ8) in the presence of 4-phenoxypyridine-derived compounds in an aqueous environment are discussed. Overall, the rectosynthesis of the designed drug inhibitors (P54 and P55) and their in vitro and in vivo bioactivity evaluation may be attractive for design and discovery of novel drug effective to inhibit c-Met enzymatic activity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ossama Daoui
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, Fez, Morocco
| | - Hassan Nour
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - Oussama Abchir
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - Souad Elkhattabi
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, Fez, Morocco
| | - Mohamed Bakhouch
- Laboratory of Bioorganic Chemistry, Department of Chemistry, Faculty of Sciences, Chouaïb Doukkali University, El Jadida, Morocco
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| |
Collapse
|
8
|
Mortazavi M, Divar M, Damghani T, Moosavi F, Saso L, Pirhadi S, Khoshneviszadeh M, Edraki N, Firuzi O. Study of the anticancer effect of new quinazolinone hydrazine derivatives as receptor tyrosine kinase inhibitors. Front Chem 2022; 10:969559. [PMID: 36465863 PMCID: PMC9713320 DOI: 10.3389/fchem.2022.969559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/18/2022] [Indexed: 09/08/2023] Open
Abstract
The advent of novel receptor tyrosine kinase inhibitors has provided an important therapeutic tool for cancer patients. In this study, a series of quinazolinone hydrazide triazole derivatives were designed and synthesized as novel MET (c-MET) receptor tyrosine kinase inhibitors. The antiproliferative effect of the synthesized compounds was examined against EBC-1, A549, HT-29 and U-87MG cells by MTT assay. MET kinase inhibitory effect was tested by a Homogenous Time Resolved Fluorescence (HTRF) assay. The antiproliferative effect of compounds in a three-dimensional spheroid culture was studied by acid phosphatase (APH) assay, while apoptosis induction was examined by Hoechst 33258 staining. We found that compound CM9 bearing p-bromo benzyl pendant inhibited MET kinase activity at the concentrations of 10-50 μM (% Inhibition = 37.1-66.3%). Compound CM9 showed antiproliferative effect against cancer cells, in particular lung cancer cells with MET amplification (EBC-1) with an IC50 value of 8.6 μM. Moreover, this derivative inhibited cell growth in spheroid cultures in a dose-dependent manner and induced apoptosis in cancer cells. Assessment of inhibitory effect of CM9 against a panel of 18 different protein kinases demonstrated that this compound also inhibits ALK, AXL, FGFR1, FLT1 (VEGFR1) and FLT4 (VEGFR3) more than 50% at 25 μM. Finally, molecular docking and dynamics simulation corroborated the experimental findings and showed critical structural features for the interactions between CM9 and target kinases. The findings of this study present quinazolinone hydrazide triazole derivatives as kinase inhibitors with considerable anticancer effects.
Collapse
Affiliation(s)
- Motahareh Mortazavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoumeh Divar
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Damghani
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Somayeh Pirhadi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Ghosh S, Cho SJ. Structure–activity
relationship and
in silico
development of
c‐Met
kinase inhibitors. B KOREAN CHEM SOC 2022. [DOI: 10.1002/bkcs.12537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Suparna Ghosh
- Department of Biomedical Sciences College of Medicine, Chosun University Gwangju Republic of Korea
| | - Seung Joo Cho
- Department of Biomedical Sciences College of Medicine, Chosun University Gwangju Republic of Korea
- Department of Cellular Molecular Medicine College of Medicine, Chosun University Gwangju Republic of Korea
| |
Collapse
|