1
|
Sima L, Wang Z, Yu L, Hou Y, Zhao D, Luo B, Liao W, Liu X. Discovery of LAH-1 as potent c-Met inhibitor for the treatment of non-small cell lung cancer. J Enzyme Inhib Med Chem 2024; 39:2286435. [PMID: 38078363 DOI: 10.1080/14756366.2023.2286435] [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: 06/16/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
ABSTRCTDysregulated HGF/c-Met pathway has been implicated in multiple human cancers and has become an attractive target for cancer intervention. Herein, we report the discovery of N-(3-fluoro-4-((2-(3-hydroxyazetidine-1-carboxamido)pyridin-4-yl)oxy)phenyl)-1-(4-fluorophenyl)-4-methyl-6-oxo-1,6-dihydropyridazine-3-carboxamide (LAH-1), which demonstrated nanomolar MET kinase activity as well as desirable antiproliferative activity, especially against EBC-1 cells. Mechanism studies confirmed the effects of LAH-1 on modulation of HGF/c-Met pathway, induction of cell apoptosis, inhibition on colony formation as well as cell migration and invasion. In addition, LAH-1 also showed desirable in vitro ADME properties as well as acceptable in vivo PK parameters. The design, synthesis, and characterisation of LAH-1 are described herein.
Collapse
Affiliation(s)
- Lijie Sima
- Department of Hematology and Oncology, The Affiliated Shaoyang Hospital, Hengyang Medical School, University of South China (Shaoyang Central Hospital), Shaoyang, China
| | - Zhongyuan Wang
- Department of Pharmacy, Guizhou Provincial People's Hospital, Guiyang, China
| | - Ling Yu
- Department of Pharmacy, Guiyang Healthcare Vocational University, Guiyang, China
| | - Youli Hou
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Dongsheng Zhao
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Bilan Luo
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Weike Liao
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Xinfu Liu
- Department of Hematology and Oncology, The Affiliated Shaoyang Hospital, Hengyang Medical School, University of South China (Shaoyang Central Hospital), Shaoyang, China
| |
Collapse
|
2
|
Ying S, Chi H, Wu X, Zeng P, Chen J, Fu T, Fu W, Zhang P, Tan W. Selective and Orally Bioavailable c-Met PROTACs for the Treatment of c-Met-Addicted Cancer. J Med Chem 2024; 67:17053-17069. [PMID: 39348183 DOI: 10.1021/acs.jmedchem.3c02417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
c-Met is an attractive therapeutic target in multiple tumors. Previous studies have discovered some effective proteolysis-targeting chimeras (PROTACs) able to degrade c-Met; however, the structure-activity relationship (SAR), degradation selectivity, and pharmacokinetic profiles of c-Met PROTACs have, to date, remained largely unknown. Herein, through extensive SAR studies on various warheads, linkers, and E3 ligase ligands, a novel potent c-Met PROTAC Met-DD4 was identified. Our results suggested that Met-DD4 could induce robust c-Met degradation with excellent selectivity (DC50 = 6.21 nM), substantially killing the c-Met-addicted cancer cells (IC50 = 4.37 nM). Furthermore, in vivo studies showed that Met-DD4 could achieve excellent oral bioavailability and c-Met degradation, strongly retarding tumor growth with minute organ toxicity. Overall, this study reveals that targeted degradation of c-Met is a promising strategy for the treatment of c-Met-addicted cancers and provides novel lead compounds for the clinical translation of c-Met PROTACs.
Collapse
Affiliation(s)
- Shilong Ying
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Hongli Chi
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xiaoqiu Wu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Pingping Zeng
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Jinling Chen
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ting Fu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Weitao Fu
- Insitute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| | - Penghui Zhang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Weihong Tan
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
3
|
Wang Y, Jia J, Wang F, Fang Y, Yang Y, Zhou Q, Yuan W, Gu X, Hu J, Yang S. Pre-metastatic niche: formation, characteristics and therapeutic implication. Signal Transduct Target Ther 2024; 9:236. [PMID: 39317708 PMCID: PMC11422510 DOI: 10.1038/s41392-024-01937-7] [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: 02/28/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 09/26/2024] Open
Abstract
Distant metastasis is a primary cause of mortality and contributes to poor surgical outcomes in cancer patients. Before the development of organ-specific metastasis, the formation of a pre-metastatic niche is pivotal in promoting the spread of cancer cells. This review delves into the intricate landscape of the pre-metastatic niche, focusing on the roles of tumor-derived secreted factors, extracellular vesicles, and circulating tumor cells in shaping the metastatic niche. The discussion encompasses cellular elements such as macrophages, neutrophils, bone marrow-derived suppressive cells, and T/B cells, in addition to molecular factors like secreted substances from tumors and extracellular vesicles, within the framework of pre-metastatic niche formation. Insights into the temporal mechanisms of pre-metastatic niche formation such as epithelial-mesenchymal transition, immunosuppression, extracellular matrix remodeling, metabolic reprogramming, vascular permeability and angiogenesis are provided. Furthermore, the landscape of pre-metastatic niche in different metastatic organs like lymph nodes, lungs, liver, brain, and bones is elucidated. Therapeutic approaches targeting the cellular and molecular components of pre-metastatic niche, as well as interventions targeting signaling pathways such as the TGF-β, VEGF, and MET pathways, are highlighted. This review aims to enhance our understanding of pre-metastatic niche dynamics and provide insights for developing effective therapeutic strategies to combat tumor metastasis.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Jiachi Jia
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Yingshuai Fang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Yabing Yang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Quanbo Zhou
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Xiaoming Gu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| |
Collapse
|
4
|
Paoletti N, Supuran CT. Benzothiazole derivatives in the design of antitumor agents. Arch Pharm (Weinheim) 2024; 357:e2400259. [PMID: 38873921 DOI: 10.1002/ardp.202400259] [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: 04/08/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024]
Abstract
Benzothiazoles are a class of heterocycles with multiple applications as anticancer, antibiotic, antiviral, and anti-inflammatory agents. Benzothiazole is a privileged scaffold in drug discovery programs for modulating a variety of biological functions. This review focuses on the design and synthesis of new benzothiazole derivatives targeting hypoxic tumors. Cancer is a major health problem, being among the leading causes of death. Tumor-hypoxic areas promote proliferation, malignancy, and resistance to drug treatment, leading to the dysregulation of key signaling pathways that involve drug targets such as vascular endothelial growth factor, epidermal growth factor receptor, hepatocyte growth factor receptor, dual-specificity protein kinase, cyclin-dependent protein kinases, casein kinase 2, Rho-related coil formation protein kinase, tunica interna endothelial cell kinase, cyclooxygenase-2, adenosine kinase, lysophosphatidic acid acyltransferases, stearoyl-CoA desaturase, peroxisome proliferator-activated receptors, thioredoxin, heat shock proteins, and carbonic anhydrase IX/XII. In turn, they regulate angiogenesis, proliferation, differentiation, and cell survival, controlling the cell cycle, inflammation, the immune system, and metabolic alterations. A wide diversity of benzothiazoles were reported over the last years to interfere with various proteins involved in tumorigenesis and, more specifically, in hypoxic tumors. Many hypoxic targets are overexpressed as a result of the hypoxia-inducible factor activation cascade and may not be present in normal tissues, providing a potential strategy for selectively targeting hypoxic cancers.
Collapse
Affiliation(s)
- Niccolò Paoletti
- Department of Neurofarba, Section of Pharmaceutical & Nutraceutical Sciences, Polo Scientifico, University of Florence, Sesto Fiorentino (Firenze), Italy
| | - Claudiu T Supuran
- Department of Neurofarba, Section of Pharmaceutical & Nutraceutical Sciences, Polo Scientifico, University of Florence, Sesto Fiorentino (Firenze), Italy
| |
Collapse
|
5
|
Wang Y, Nan X, Duan Y, Wang Q, Liang Z, Yin H. FDA-approved small molecule kinase inhibitors for cancer treatment (2001-2015): Medical indication, structural optimization, and binding mode Part I. Bioorg Med Chem 2024; 111:117870. [PMID: 39128361 DOI: 10.1016/j.bmc.2024.117870] [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: 07/03/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
The dysregulation of kinases has emerged as a major class of targets for anticancer drug discovery given its node roles in the etiology of tumorigenesis, progression, invasion, and metastasis of malignancies, which is validated by the FDA approval of 28 small molecule kinase inhibitor (SMKI) drugs for cancer treatment at the end of 2015. While the preclinical and clinical data of these drugs are widely presented, it is highly essential to give an updated review on the medical indications, design principles and binding modes of these anti-tumor SMKIs approved by the FDA to offer insights for the future development of SMKIs with specific efficacy and safety.
Collapse
Affiliation(s)
- Ying Wang
- Department of Electrophysiological Diagnosis, 3201 Hospital of Xi'an Jiaotong University Health Science Center, Hanzhong 723000, China
| | - Xiang Nan
- College of Chemical & Environment Science, Shaanxi University of Technology, Hanzhong 723001, China; Department of Stomatology, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Yanping Duan
- College of Chemical & Environment Science, Shaanxi University of Technology, Hanzhong 723001, China
| | - Qiuxu Wang
- Department of Stomatology, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| | - Zhigang Liang
- Department of Stomatology, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Hanrong Yin
- Department of Electrophysiological Diagnosis, 3201 Hospital of Xi'an Jiaotong University Health Science Center, Hanzhong 723000, China.
| |
Collapse
|
6
|
Yao S, Liu X, Feng Y, Li Y, Xiao X, Han Y, Xia S. Unveiling the Role of HGF/c-Met Signaling in Non-Small Cell Lung Cancer Tumor Microenvironment. Int J Mol Sci 2024; 25:9101. [PMID: 39201787 PMCID: PMC11354629 DOI: 10.3390/ijms25169101] [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: 07/28/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is characterized by several molecular alterations that contribute to its development and progression. These alterations include the epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), human epidermal growth factor receptor 2 (HER2), and mesenchymal-epithelial transition factor (c-MET). Among these, the hepatocyte growth factor (HGF)/c-MET signaling pathway plays a crucial role in NSCLC. In spite of this, the involvement of the HGF/c-MET signaling axis in remodeling the tumor microenvironment (TME) remains relatively unexplored. This review explores the biological functions of the HGF/c-MET signaling pathway in both normal and cancerous cells, examining its multifaceted roles in the NSCLC tumor microenvironment, including tumor cell proliferation, migration and invasion, angiogenesis, and immune evasion. Furthermore, we summarize the current progress and clinical applications of MET-targeted therapies in NSCLC and discuss future research directions, such as the development of novel MET inhibitors and the potential of combination immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shu Xia
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (S.Y.); (X.L.); (Y.F.); (Y.L.); (X.X.); (Y.H.)
| |
Collapse
|
7
|
Estevam GO, Linossi EM, Rao J, Macdonald CB, Ravikumar A, Chrispens KM, Capra JA, Coyote-Maestas W, Pimentel H, Collisson EA, Jura N, Fraser JS. Mapping kinase domain resistance mechanisms for the MET receptor tyrosine kinase via deep mutational scanning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603579. [PMID: 39071407 PMCID: PMC11275805 DOI: 10.1101/2024.07.16.603579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Mutations in the kinase and juxtamembrane domains of the MET Receptor Tyrosine Kinase are responsible for oncogenesis in various cancers and can drive resistance to MET-directed treatments. Determining the most effective inhibitor for each mutational profile is a major challenge for MET-driven cancer treatment in precision medicine. Here, we used a deep mutational scan (DMS) of ~5,764 MET kinase domain variants to profile the growth of each mutation against a panel of 11 inhibitors that are reported to target the MET kinase domain. We identified common resistance sites across type I, type II, and type I ½ inhibitors, unveiled unique resistance and sensitizing mutations for each inhibitor, and validated non-cross-resistant sensitivities for type I and type II inhibitor pairs. We augment a protein language model with biophysical and chemical features to improve the predictive performance for inhibitor-treated datasets. Together, our study demonstrates a pooled experimental pipeline for identifying resistance mutations, provides a reference dictionary for mutations that are sensitized to specific therapies, and offers insights for future drug development.
Collapse
Affiliation(s)
- Gabriella O. Estevam
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
- Tetrad Graduate Program, UCSF, San Francisco, CA, United States
| | - Edmond M. Linossi
- Cardiovascular Research Institute, UCSF, San Francisco, CA, United States
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, United States
| | - Jingyou Rao
- Department of Computer Science, UCLA, Los Angeles, CA, United States
| | - Christian B. Macdonald
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
| | - Ashraya Ravikumar
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
| | - Karson M. Chrispens
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
- Biophysics Graduate Program, UCSF, San Francisco, CA, United States
| | - John A. Capra
- Bakar Computational Health Sciences Institute and Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, United States
| | - Willow Coyote-Maestas
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
- Quantitative Biosciences Institute, UCSF, San Francisco, CA, United States
| | - Harold Pimentel
- Department of Computer Science, UCLA, Los Angeles, CA, United States
- Department of Computational Medicine and Human Genetics, UCLA, Los Angeles, CA, United States
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Eric A. Collisson
- Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States
- Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Natalia Jura
- Cardiovascular Research Institute, UCSF, San Francisco, CA, United States
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, United States
- Quantitative Biosciences Institute, UCSF, San Francisco, CA, United States
| | - James S. Fraser
- Department of Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, United States
- Quantitative Biosciences Institute, UCSF, San Francisco, CA, United States
| |
Collapse
|
8
|
Zhang Y, Ma M, Yang J, Qiu X, Xin L, Lu Y, Huang H, Zeng Z, Zeng D. Preparation, Characterization, and Oral Bioavailability of Solid Dispersions of Cryptosporidium parvum Alternative Oxidase Inhibitors. Int J Mol Sci 2024; 25:7025. [PMID: 39000132 PMCID: PMC11241238 DOI: 10.3390/ijms25137025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/12/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
The phenylpyrazole derivative 5-amino-3-[1-cyano-2-(3-phenyl-1H-pyrazol-4-yl) vinyl]-1-phenyl-1H-pyrazole-4-carbonitrile (LN002), which was screened out through high-throughput molecular docking for the AOX target, exhibits promising efficacy against Cryptosporidium. However, its poor water solubility limits its oral bioavailability and therapeutic utility. In this study, solid dispersion agents were prepared by using HP-β-CD and Soluplus® and characterized through differential scanning calorimetry, Fourier transform infrared, powder X-ray diffraction, and scanning electron microscopy. Physical and chemical characterization showed that the crystal morphology of LN002 transformed into an amorphous state, thus forming a solid dispersion of LN002. The solid dispersion prepared with an LN002/HP-β-CD/Soluplus® mass ratio of 1:3:9 (w/w/w) exhibited significantly increased solubility and cumulative dissolution. Meanwhile, LN002 SDs showed good preservation stability under accelerated conditions of 25 °C and 75% relative humidity. The complexation of LN002 with HP-β-CD and Soluplus® significantly improved water solubility, pharmacological properties, absorption, and bioavailability.
Collapse
Affiliation(s)
- Yongxiang Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou 510642, China
| | - Minglang Ma
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou 510642, China
| | - Jinyu Yang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou 510642, China
| | - Xiaotong Qiu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou 510642, China
| | - Lin Xin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou 510642, China
| | - Yixing Lu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou 510642, China
| | - Huiguo Huang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou 510642, China
| | - Zhenling Zeng
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou 510642, China
| | - Dongping Zeng
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, Guangzhou 510642, China
| |
Collapse
|
9
|
Thu YM, Suzawa K, Tomida S, Ochi K, Tsudaka S, Takatsu F, Date K, Matsuda N, Iwata K, Nakata K, Shien K, Yamamoto H, Okazaki M, Sugimoto S, Toyooka S. PAI-1 mediates acquired resistance to MET-targeted therapy in non-small cell lung cancer. PLoS One 2024; 19:e0300644. [PMID: 38758826 PMCID: PMC11101109 DOI: 10.1371/journal.pone.0300644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/03/2024] [Indexed: 05/19/2024] Open
Abstract
Mechanisms underlying primary and acquired resistance to MET tyrosine kinase inhibitors (TKIs) in managing non-small cell lung cancer remain unclear. In this study, we investigated the possible mechanisms acquired for crizotinib in MET-amplified lung carcinoma cell lines. Two MET-amplified lung cancer cell lines, EBC-1 and H1993, were established for acquired resistance to MET-TKI crizotinib and were functionally elucidated. Genomic and transcriptomic data were used to assess the factors contributing to the resistance mechanism, and the alterations hypothesized to confer resistance were validated. Multiple mechanisms underlie acquired resistance to crizotinib in MET-amplified lung cancer cell lines. In EBC-1-derived resistant cells, the overexpression of SERPINE1, the gene encoding plasminogen activator inhibitor-1 (PAI-1), mediated the drug resistance mechanism. Crizotinib resistance was addressed by combination therapy with a PAI-1 inhibitor and PAI-1 knockdown. Another mechanism of resistance in different subline cells of EBC-1 was evaluated as epithelial-to-mesenchymal transition with the upregulation of antiapoptotic proteins. In H1993-derived resistant cells, MEK inhibitors could be a potential therapeutic strategy for overcoming resistance with downstream mitogen-activated protein kinase pathway activation. In this study, we revealed the different mechanisms of acquired resistance to the MET inhibitor crizotinib with potential therapeutic application in patients with MET-amplified lung carcinoma.
Collapse
Affiliation(s)
- Yin Min Thu
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ken Suzawa
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shuta Tomida
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Kosuke Ochi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shimpei Tsudaka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Fumiaki Takatsu
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keiichi Date
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Naoki Matsuda
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuma Iwata
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kentaro Nakata
- Department of Surgery, Division of Cardiovascular and Thoracic Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Kazuhiko Shien
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromasa Yamamoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mikio Okazaki
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
10
|
Fang MM, Cheng JT, Chen YQ, Lin XC, Su JW, Wu YL, Chen HJ, Yang JJ. Molecular features and clinical outcomes of EGFR-mutated, MET-amplified non-small-cell lung cancer after resistance to dual-targeted therapy. Ther Adv Med Oncol 2024; 16:17588359241234504. [PMID: 38449561 PMCID: PMC10916476 DOI: 10.1177/17588359241234504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/30/2024] [Indexed: 03/08/2024] Open
Abstract
Background Some studies of dual-targeted therapy (DTT) targeting epidermal growth factor receptor (EGFR) and mesenchymal-epithelial transition (MET) have shown promising efficacy in non-small-cell lung cancer (NSCLC). Consequently, patient management following DTT resistance has gained significance. However, the underlying resistance mechanisms and clinical outcomes in these patients remain unclear. Objectives This study aimed to delineate the molecular characteristics and survival outcomes of patients with NSCLC harboring EGFR mutations and acquired MET amplification after developing resistance to DTT. Design We conducted a retrospective analysis of patients with NSCLC with EGFR mutations and acquired MET amplification who exhibited resistance to EGFR/MET DTT. Methods Next-generation sequencing (NGS) was performed on patients with available tissue samples before and/or after the development of resistance to DTT. Stratified analyses were carried out based on data sources and subsequent salvage treatments. Univariate/multivariate Cox regression models and survival analyses were employed to explore potential independent prognostic factors. Results The study included 77 NSCLC patients, with NGS conducted on 19 patients. We observed many resistance mechanisms, including EGFR-dependent pathways (4/19, 21.1%), MET-dependent pathways (2/19, 10.5%), EGFR/MET co-dependent pathways (2/19, 10.5%), and EGFR/MET-independent resistance mechanisms (11/19, 57.9%). Post-progression progression-free survival (pPFS) and post-progression overall survival (pOS) significantly varied among patients who received the best supportive care (BSC), targeted therapy, or chemotherapy (CT), with median pPFS of 1.5, 3.9, and 4.9 months, respectively (p = 0.003). Median pOS were 2.3, 7.7, and 9.2 months, respectively (p < 0.001). The number of treatment lines following DTT resistance and the Eastern Cooperative Oncology Group performance status emerged as the independent prognostic factors. Conclusion This study revealed a heterogeneous landscape of resistance mechanisms to EGFR/MET DTT, with a similar prevalence of on- and off-target mechanisms. Targeted therapy or CT, as compared to BSC, exhibited the potential to improve survival outcomes for patients with advanced NSCLC following resistance to DTT.
Collapse
Affiliation(s)
- Mei-Mei Fang
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiang-Tao Cheng
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yu-Qing Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiao-Cheng Lin
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jun-Wei Su
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hua-Jun Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou 510080, China
| |
Collapse
|
11
|
Lien VT, Hauge E, Nuruddin S, Klaveness J, Olberg DE. Synthesis and preclinical evaluation of a selective MET kinase positron emission tomography tracer. J Labelled Comp Radiopharm 2023; 66:452-460. [PMID: 37867318 DOI: 10.1002/jlcr.4066] [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: 03/15/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 10/24/2023]
Abstract
The tyrosine kinase MET (hepatocyte growth factor receptor) is activated or mutated in a wide range of cancers and is often correlated with a poor prognosis. Precision medicine with positron emission tomography (PET) can potentially aid in the assessment of tumor biochemistry and heterogeneity, which can prompt the selection of the most effective therapeutic regimes. The selective MET inhibitor PF04217903 (1) formed the basis for a bioisosteric replacement, leading to the deoxyfluorinated analog [18 F]2. [18 F]2 could be synthesized with a "hydrous fluoroethylation" protocol in 6.3 ± 2.6% radiochemical yield and a molar activity of >50 GBq/μmol. In vitro autoradiography indicated that [18 F]2 selectively binds to MET in PC3 tumor tissue, and in vivo biodistribution in mice showed predominantly a hepatobiliary excretion along with a low retention of radiotracer in other organs.
Collapse
Affiliation(s)
- Vegard Torp Lien
- Department of Pharmacy, University of Oslo, Oslo, Norway
- Norwegian Medical Cyclotron Center, Oslo, Norway
| | - Emily Hauge
- Department of Pharmacy, University of Oslo, Oslo, Norway
- Norwegian Medical Cyclotron Center, Oslo, Norway
| | | | - Jo Klaveness
- Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Dag Erlend Olberg
- Department of Pharmacy, University of Oslo, Oslo, Norway
- Norwegian Medical Cyclotron Center, Oslo, Norway
| |
Collapse
|
12
|
Nan X, Wang QX, Xing SJ, Liang ZG. Design, synthesis, and biological evaluation of thiazole/thiadiazole carboxamide scaffold-based derivatives as potential c-Met kinase inhibitors for cancer treatment. J Enzyme Inhib Med Chem 2023; 38:2247183. [PMID: 37642355 PMCID: PMC10467532 DOI: 10.1080/14756366.2023.2247183] [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: 05/11/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/31/2023] Open
Abstract
As part of our continuous efforts to discover novel c-Met inhibitors as antitumor agents, four series of thiazole/thiadiazole carboxamide-derived analogues were designed, synthesised, and evaluated for the in vitro activity against c-Met and four human cancer cell lines. After five cycles of optimisation on structure-activity relationship, compound 51am was found to be the most promising inhibitor in both biochemical and cellular assays. Moreover, 51am exhibited potency against several c-Met mutants. Mechanistically, 51am not only induced cell cycle arrest and apoptosis in MKN-45 cells but also inhibited c-Met phosphorylation in the cell and cell-free systems. It also exhibited a good pharmacokinetic profile in BALB/c mice. Furthermore, the binding mode of 51am with both c-Met and VEGFR-2 provided novel insights for the discovery of selective c-Met inhibitors. Taken together, these results indicate that 51am could be an antitumor candidate meriting further development.
Collapse
Affiliation(s)
- Xiang Nan
- Department of Stomatology, Shenzhen Second People’s Hospital, Shenzhen, China
- School of Biomedical Engineering, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Shenzhen University Medical School, Shenzhen, China
| | - Qiu-Xu Wang
- Department of Stomatology, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Shao-Jun Xing
- School of Biomedical Engineering, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Shenzhen University Medical School, Shenzhen, China
| | - Zhi-Gang Liang
- Department of Stomatology, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
13
|
Grädler U, Schwarz D, Wegener A, Eichhorn T, Bandeiras TM, Freitas MC, Lammens A, Ganichkin O, Augustin M, Minguzzi S, Becker F, Bomke J. Biophysical and structural characterization of the impacts of MET phosphorylation on tepotinib binding. J Biol Chem 2023; 299:105328. [PMID: 37806493 PMCID: PMC10654029 DOI: 10.1016/j.jbc.2023.105328] [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: 01/24/2023] [Revised: 08/16/2023] [Accepted: 09/07/2023] [Indexed: 10/10/2023] Open
Abstract
The receptor tyrosine kinase MET is activated by hepatocyte growth factor binding, followed by phosphorylation of the intracellular kinase domain (KD) mainly within the activation loop (A-loop) on Y1234 and Y1235. Dysregulation of MET can lead to both tumor growth and metastatic progression of cancer cells. Tepotinib is a highly selective, potent type Ib MET inhibitor and approved for treatment of non-small cell lung cancer harboring METex14 skipping alterations. Tepotinib binds to the ATP site of unphosphorylated MET with critical π-stacking contacts to Y1230 of the A-loop, resulting in a high residence time. In our study, we combined protein crystallography, biophysical methods (surface plasmon resonance, differential scanning fluorimetry), and mass spectrometry to clarify the impacts of A-loop conformation on tepotinib binding using different recombinant MET KD protein variants. We solved the first crystal structures of MET mutants Y1235D, Y1234E/1235E, and F1200I in complex with tepotinib. Our biophysical and structural data indicated a linkage between reduced residence times for tepotinib and modulation of A-loop conformation either by mutation (Y1235D), by affecting the overall Y1234/Y1235 phosphorylation status (L1195V and F1200I) or by disturbing critical π-stacking interactions with tepotinib (Y1230C). We corroborated these data with target engagement studies by fluorescence cross-correlation spectroscopy using KD constructs in cell lysates or full-length receptors from solubilized cellular membranes as WT or activated mutants (Y1235D and Y1234E/1235E). Collectively, our results provide further insight into the MET A-loop structural determinants that affect the binding of the selective inhibitor tepotinib.
Collapse
Affiliation(s)
- Ulrich Grädler
- The Healthcare Business of Merck KGaA, Darmstadt, Germany.
| | - Daniel Schwarz
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Ansgar Wegener
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | - Tiago M Bandeiras
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Micael C Freitas
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | | | | | | | | | | | - Jörg Bomke
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| |
Collapse
|
14
|
Alzain AA, Elbadwi FA, Mohamed SGA, Kushk KSA, Bafarhan RI, Alswiri SA, Khushaim SN, Hussein HGA, Abuhajras MYA, Mohamed GA, Ibrahim SRM. Exploring marine-derived compounds for MET signalling pathway inhibition in cancer: integrating virtual screening, ADME profiling and molecular dynamics investigations. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2023; 34:1003-1021. [PMID: 38014514 DOI: 10.1080/1062936x.2023.2284917] [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: 08/28/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023]
Abstract
The MET signalling pathway regulates fundamental cellular processes such as growth, division, and survival. While essential for normal cell function, dysregulation of this pathway can contribute to cancer by triggering uncontrolled proliferation and metastasis. Targeting MET activity holds promise as an effective strategy for cancer therapy. Among potential sources of anti-cancer agents, marine organisms have gained attention. In this study, we screened 47,450 natural compounds derived from marine sources within the CMNPD database against the Met crystal structure. By employing HTVS, SP, and XP docking modes, we identified three compounds (CMNPD17595, CMNPD14026, and CMNPD19696) that outperformed a reference molecule in binding affinity to the Met structure. These compounds demonstrated desirable ADME properties. Molecular Dynamics (MD) simulations for 200 ns confirmed the stability of their interactions with Met. Our findings highlight CMNPD17595, CMNPD14026, and CMNPD19696 as potential inhibitors against Met-dependent cancers. Additionally, these compounds offer new avenues for drug development, leveraging their inhibitory effects on Met to combat carcinogenesis.
Collapse
Affiliation(s)
- A A Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - F A Elbadwi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - S G A Mohamed
- Faculty of Dentistry, British University, El Sherouk City, Egypt
| | - K S A Kushk
- Operations Sales Department, United Pharmaceuticals & Medical Supply Co. Ltd, Al Madinah Al-Munawwarah, Saudi Arabia
| | - R I Bafarhan
- Pharmaceutical Care Services, Medical Department, Private Sector, Tabuk, Saudi Arabia
| | - S A Alswiri
- Pharmaceutical Company, Medical Department, Private Sector, Al Madinah Al-Munawwarah, Saudi Arabia
| | - S N Khushaim
- College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - H G A Hussein
- Preparatory Year Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - M Y A Abuhajras
- Medical Claims Department, Bupa Arabia, Prince Saud AlFaisal, Jeddah, Saudi Arabia
| | - G A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - S R M Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
15
|
Spitaleri G, Trillo Aliaga P, Attili I, Del Signore E, Corvaja C, Corti C, Uliano J, Passaro A, de Marinis F. MET in Non-Small-Cell Lung Cancer (NSCLC): Cross 'a Long and Winding Road' Looking for a Target. Cancers (Basel) 2023; 15:4779. [PMID: 37835473 PMCID: PMC10571577 DOI: 10.3390/cancers15194779] [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: 08/11/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Non-Small-Cell Lung Cancer (NSCLC) can harbour different MET alterations, such as MET overexpression (MET OE), MET gene amplification (MET AMP), or MET gene mutations. Retrospective studies of surgical series of patients with MET-dysregulated NSCLC have shown worse clinical outcomes irrespective of the type of specific MET gene alteration. On the other hand, earlier attempts failed to identify the 'druggable' molecular gene driver until the discovery of MET exon 14 skipping mutations (METex14). METex14 are rare and amount to around 3% of all NSCLCs. Patients with METex14 NSCLC attain modest results when they are treated with immune checkpoint inhibitors (ICIs). New selective MET inhibitors (MET-Is) showed a long-lasting clinical benefit in patients with METex14 NSCLC and modest activity in patients with MET AMP NSCLC. Ongoing clinical trials are investigating new small molecule tyrosine kinase inhibitors, bispecific antibodies, or antibodies drug conjugate (ADCs). This review focuses on the prognostic role of MET, the summary of pivotal clinical trials of selective MET-Is with a focus on resistance mechanisms. The last section is addressed to future developments and challenges.
Collapse
Affiliation(s)
- Gianluca Spitaleri
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Pamela Trillo Aliaga
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Ilaria Attili
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Ester Del Signore
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Carla Corvaja
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (C.C.); (J.U.)
- Department of Oncology and Haematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Jacopo Uliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (C.C.); (J.U.)
- Department of Oncology and Haematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| | - Filippo de Marinis
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (P.T.A.); (I.A.); (E.D.S.); (C.C.); (F.d.M.)
| |
Collapse
|
16
|
Nan X, Li X, Wu Y, Li H, Wang Q, Xing S, Liang Z. Design, synthesis and biological evaluation of sulfonylamidines as potent c-Met inhibitors by enhancing hydrophobic interaction. Org Biomol Chem 2023; 21:7459-7466. [PMID: 37667983 DOI: 10.1039/d3ob01156a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
The dysregulation of c-Met kinase has emerged as a significant contributing factor for the occurrence, progression, poor clinical outcomes and drug resistance of various human cancers. In our ongoing pursuit to identify promising c-Met inhibitors as potential antitumor agents, a docking study of the previously reported c-Met inhibitor 7 revealed a large unoccupied hydrophobic pocket, which could present an opportunity for further exploration of structure-activity relationships to improve the binding affinity with the allosteric hydrophobic back pocket of c-Met. Herein we performed structure-activity relationship and molecular modeling studies based on lead compound 7. The collective endeavors culminated in the discovery of compound 21j with superior efficacy to 7 and positive control foretinib by increasing the hydrophobic interaction with the hydrophobic back pocket of c-Met active site.
Collapse
Affiliation(s)
- Xiang Nan
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
- Department of Stomatology, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| | - Xin Li
- Weihai Marine Organism & Medical Technology Research Institute, Harbin Institute of Technology, Weihai 264209, China.
| | - Yanchao Wu
- Weihai Marine Organism & Medical Technology Research Institute, Harbin Institute of Technology, Weihai 264209, China.
| | - Huijing Li
- Weihai Marine Organism & Medical Technology Research Institute, Harbin Institute of Technology, Weihai 264209, China.
| | - Qiuxu Wang
- Department of Stomatology, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| | - Shaojun Xing
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Zhigang Liang
- Department of Stomatology, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| |
Collapse
|
17
|
Hsu R, Benjamin DJ, Nagasaka M. The Development and Role of Capmatinib in the Treatment of MET-Dysregulated Non-Small Cell Lung Cancer-A Narrative Review. Cancers (Basel) 2023; 15:3561. [PMID: 37509224 PMCID: PMC10377299 DOI: 10.3390/cancers15143561] [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: 05/24/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of death, but over the past decade, there has been tremendous progress in the field with new targeted therapies. The mesenchymal-epithelial transition factor (MET) proto-oncogene has been implicated in multiple solid tumors, including NSCLC, and dysregulation in NSCLC from MET can present most notably as MET exon 14 skipping mutation and amplification. From this, MET tyrosine kinase inhibitors (TKIs) have been developed to treat this dysregulation despite challenges with efficacy and reliable biomarkers. Capmatinib is a Type Ib MET TKI first discovered in 2011 and was FDA approved in August 2022 for advanced NSCLC with MET exon 14 skipping mutation. In this narrative review, we discuss preclinical and early-phase studies that led to the GEOMETRY mono-1 study, which showed beneficial efficacy in MET exon 14 skipping mutations, leading to FDA approval of capmatinib along with Foundation One CDx assay as its companion diagnostic assay. Current and future directions of capmatinib are focused on improving the efficacy, overcoming the resistance of capmatinib, and finding approaches for new indications of capmatinib such as acquired MET amplification from epidermal growth factor receptor (EGFR) TKI resistance. Clinical trials now involve combination therapy with capmatinib, including amivantamab, trametinib, and immunotherapy. Furthermore, new drug agents, particularly antibody-drug conjugates, are being developed to help treat patients with acquired resistance from capmatinib and other TKIs.
Collapse
Affiliation(s)
- Robert Hsu
- Division of Medical Oncology, Department of Internal Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | | | - Misako Nagasaka
- Division of Hematology and Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA 92868, USA
| |
Collapse
|
18
|
Wang C, Lu X. Targeting MET: Discovery of Small Molecule Inhibitors as Non-Small Cell Lung Cancer Therapy. J Med Chem 2023. [PMID: 37262349 DOI: 10.1021/acs.jmedchem.3c00028] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
MET has been considered as a promising drug target for the treatment of MET-dependent diseases, particularly non-small cell lung cancer (NSCLC). Small molecule MET inhibitors with mainly three types of binding modes (Ia/Ib, II, and III) have been developed. In this Review, we provide an overview of the structural features, activation mechanism, and dysregulation pathway of MET and summarize progress on the development and discovery strategies utilized for MET inhibitors as well as mechanisms of acquired resistance to current approved inhibitors. The insights will accelerate discovery of new generation MET inhibitors to overcome clinical acquired resistance.
Collapse
Affiliation(s)
- Chaofan Wang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 510632, China
| | - Xiaoyun Lu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450001, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 510632, China
| |
Collapse
|
19
|
Huang G, Cierpicki T, Grembecka J. 2-Aminobenzothiazoles in anticancer drug design and discovery. Bioorg Chem 2023; 135:106477. [PMID: 36989736 PMCID: PMC10718064 DOI: 10.1016/j.bioorg.2023.106477] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/26/2023] [Accepted: 03/10/2023] [Indexed: 03/22/2023]
Abstract
Cancer is one of the major causes of mortality and morbidity worldwide. Substantial research efforts have been made to develop new chemical entities with improved anticancer efficacy. 2-Aminobenzothiazole is an important class of heterocycles containing one sulfur and two nitrogen atoms, which is associated with a broad spectrum of medical and pharmacological activities, including antitumor, antibacterial, antimalarial, anti-inflammatory, and antiviral activities. In recent years, an extraordinary collection of potent and low-toxicity 2-aminobenzothiazole compounds have been discovered as new anticancer agents. Herein, we provide a comprehensive review of this class of compounds based on their activities against tumor-related proteins, including tyrosine kinases (CSF1R, EGFR, VEGFR-2, FAK, and MET), serine/threonine kinases (Aurora, CDK, CK, RAF, and DYRK2), PI3K kinase, BCL-XL, HSP90, mutant p53 protein, DNA topoisomerase, HDAC, NSD1, LSD1, FTO, mPGES-1, SCD, hCA IX/XII, and CXCR. In addition, the anticancer potentials of 2-aminobenzothiazole-derived chelators and metal complexes are also described here. Moreover, the design strategies, mechanism of actions, structure-activity relationships (SAR) and more advanced stages of pre-clinical development of 2-aminobenzothiazoles as new anticancer agents are extensively reviewed in this article. Finally, the examples that 2-aminobenzothiazoles showcase an advantage over other heterocyclic systems are also highlighted.
Collapse
Affiliation(s)
- Guang Huang
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
20
|
Leone GM, Candido S, Lavoro A, Vivarelli S, Gattuso G, Calina D, Libra M, Falzone L. Clinical Relevance of Targeted Therapy and Immune-Checkpoint Inhibition in Lung Cancer. Pharmaceutics 2023; 15:1252. [PMID: 37111737 PMCID: PMC10142433 DOI: 10.3390/pharmaceutics15041252] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Lung cancer (LC) represents the second most diagnosed tumor and the malignancy with the highest mortality rate. In recent years, tremendous progress has been made in the treatment of this tumor thanks to the discovery, testing, and clinical approval of novel therapeutic approaches. Firstly, targeted therapies aimed at inhibiting specific mutated tyrosine kinases or downstream factors were approved in clinical practice. Secondly, immunotherapy inducing the reactivation of the immune system to efficiently eliminate LC cells has been approved. This review describes in depth both current and ongoing clinical studies, which allowed the approval of targeted therapies and immune-checkpoint inhibitors as standard of care for LC. Moreover, the present advantages and pitfalls of new therapeutic approaches will be discussed. Finally, the acquired importance of human microbiota as a novel source of LC biomarkers, as well as therapeutic targets to improve the efficacy of available therapies, was analyzed. Therapy against LC is increasingly becoming holistic, taking into consideration not only the genetic landscape of the tumor, but also the immune background and other individual variables, such as patient-specific gut microbial composition. On these bases, in the future, the research milestones reached will allow clinicians to treat LC patients with tailored approaches.
Collapse
Affiliation(s)
- Gian Marco Leone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy
| | - Alessandro Lavoro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Silvia Vivarelli
- Department of Biomedical and Dental Sciences, Morphological and Functional Imaging, Section of Occupational Medicine, University of Messina, 98125 Messina, Italy
| | - Giuseppe Gattuso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy
| | - Luca Falzone
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy;
| |
Collapse
|
21
|
Xia Y, Jin R, Li M, Lan F, Zhu H, Yu Y, Miao D, Wang Q, Zhou Y, Selvaggi G, Ying S, Zhang J, Shen H, Le X, Li W. Potent antitumor activity of ensartinib in MET exon 14 skipping-mutated non-small cell lung cancer. Cancer Lett 2023; 561:216140. [PMID: 36948240 DOI: 10.1016/j.canlet.2023.216140] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/09/2023] [Accepted: 03/19/2023] [Indexed: 03/24/2023]
Abstract
Met proto-oncogene exon 14 skipping (METex14) mutations are targetable driver genes in approximately 3% of non-small-cell lung cancers (NSCLCs). Ensartinib, a type Ia MET inhibitor, is a multi-kinase inhibitor that has been approved for ALK-positive NSCLCs. Ensartinib was administered for compassionate use (cohort 1) and in a phase II clinical trial (cohort 2) to patients with METex14 mutant NSCLCs, with ORR as a primary endpoint. Molecular simulation was conducted to evaluate ensartinib c-MET interaction, and cell lines, patient-derived organoids (PDOs), and xenograft models were used to test the effectiveness of ensartinib. Among 29 evaluable patients, the ORR and DCR of ensartinib were 67% and 94% in cohort 1, and 73% and 91% in cohort 2. The median DoR was 6.8 months and median PFS was 6.1 months in the total population. Rash was the most common drug-related adverse event, and peripheral edema of any grade was reported in only 9% patients. Molecular simulations indicated favorable binding of ensartinib to c-MET. The kinase assay demonstrated an IC50 of 7.9 nM of ensartinib against METex14 protein. In vitro, Hs746T (METex14 mutation) and EBC-1 (MET amplification) cells were sensitive to ensartinib, with IC50 of 31 and 44 nM, respectively. Ensartinib exhibited comparable inhibitory effects on cell migration as crizotinib and tepotinib in both cell types. In vivo, ensartinib suppressed the growth of Hs746T cells. Ensartinib also potently inhibited the viability of PDOs. Overall, Ensartinib exhibited substantial antitumor effects against METex14 mutant NSCLCs in preclinical and clinical trials, with relatively low peripheral edema rates.
Collapse
Affiliation(s)
- Yang Xia
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Rui Jin
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Miao Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Fen Lan
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Hao Zhu
- Department of Respiratory and Critical Care Medicine, Wuyi First People's Hospital, Jinhua, Zhejiang, China
| | - Yinghui Yu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Da Miao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiyuan Wang
- Department of Radiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Zhou
- Xcovery Holdings Inc, Palm Beach Gardens, FL, USA
| | | | - Songmin Ying
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Huahao Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA.
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
22
|
Molecular and Biological Investigation of Isolated Marine Fungal Metabolites as Anticancer Agents: A Multi-Target Approach. Metabolites 2023; 13:metabo13020162. [PMID: 36837781 PMCID: PMC9964656 DOI: 10.3390/metabo13020162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Cancer is the leading cause of death globally, with an increasing number of cases being annually reported. Nature-derived metabolites have been widely studied for their potential programmed necrosis, cytotoxicity, and anti-proliferation leading to enrichment for the modern medicine, particularly within the last couple of decades. At a more rapid pace, the concept of multi-target agents has evolved from being an innovative approach into a regular drug development procedure for hampering the multi-fashioned pathophysiology and high-resistance nature of cancer cells. With the advent of the Red Sea Penicillium chrysogenum strain S003-isolated indole-based alkaloids, we thoroughly investigated the molecular aspects for three major metabolites: meleagrin (MEL), roquefortine C (ROC), and isoroquefortine C (ISO) against three cancer-associated biological targets Cdc-25A, PTP-1B, and c-Met kinase. The study presented, for the first time, the detailed molecular insights and near-physiological affinity for these marine indole alkaloids against the assign targets through molecular docking-coupled all-atom dynamic simulation analysis. Findings highlighted the superiority of MEL's binding affinity/stability being quite in concordance with the in vitro anticancer activity profile conducted via sulforhodamine B bioassay on different cancerous cell lines reaching down to low micromolar or even nanomolar potencies. The advent of lengthy structural topologies via the metabolites' extended tetracyclic cores and aromatic imidazole arm permitted multi-pocket accommodation addressing the selectivity concerns. Additionally, the presence decorating polar functionalities on the core hydrophobic tetracyclic ring contributed compound's pharmacodynamic preferentiality. Introducing ionizable functionality with more lipophilic characters was highlighted to improve binding affinities which was also in concordance with the conducted drug-likeness/pharmacokinetic profiling for obtaining a balanced pharmacokinetic/dynamic profile. Our study adds to the knowledge regarding drug development and optimization of marine-isolated indole-based alkaloids for future iterative synthesis and pre-clinical investigations as multi-target anticancer agents.
Collapse
|
23
|
Liu H, Li X, Cai J, Jiang L, Zhang X, Wu D, Wang L, Yang A, Guo C, Chen J, Pu W, Yu F. A screening of inhibitors targeting the receptor kinase FERONIA reveals small molecules that enhance plant root immunity. PLANT BIOTECHNOLOGY JOURNAL 2023; 21:63-77. [PMID: 36121304 PMCID: PMC9829398 DOI: 10.1111/pbi.13925] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 08/24/2022] [Accepted: 09/08/2022] [Indexed: 05/14/2023]
Abstract
Receptor-like kinases (RLKs) constitute the largest receptor family involved in the regulation of plant immunity and growth, but small-molecule inhibitors that target RLKs to improve agronomic traits remain unexplored. The RLK member FERONIA (FER) negatively regulates plant resistance to certain soil-borne diseases that are difficult to control and cause huge losses in crop yields and economy. Here, we identified 33 highly effective FER kinase inhibitors from 1494 small molecules by monitoring FER autophosphorylation in vitro. Four representative inhibitors (reversine, cenisertib, staurosporine and lavendustin A) inhibited the kinase activity of FER and its homologues in several crops by targeting the conserved ATP pocket in the kinase structure. FER contributes to the physiological impact of representative inhibitors in plants. The treatment of roots with reversine, staurosporine and lavendustin A enhanced innate immunity in plant roots and thus alleviated soil-borne diseases in tobacco, tomato and rice without growth penalties. Consistently, RNA sequencing assays showed that lavendustin A and reversine exert profound impacts on immunity-related gene expression. Our results will set a new milestone in the development of the plant RLK kinase regulation theory and provide a novel strategy for the prevention and control of plant soil-borne diseases without growth penalties.
Collapse
Affiliation(s)
- Hong‐Bin Liu
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Biology, Hunan UniversityChangshaChina
| | - Xiaoxu Li
- Technology CenterChina Tobacco Hunan Industrial Co., Ltd.ChangshaChina
| | - Jun Cai
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Biology, Hunan UniversityChangshaChina
| | - Ling‐Li Jiang
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Biology, Hunan UniversityChangshaChina
| | - Xin Zhang
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life SciencesHenan UniversityKaifengChina
| | - Dousheng Wu
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Biology, Hunan UniversityChangshaChina
| | - Lifeng Wang
- State key Laboratory of Hybrid Rice, Hunan Agricultural Biotechnology Research InstituteHunan Academy of Agricultural SciencesChangshaChina
| | - Aiguo Yang
- Key Laboratory for Tobacco Gene Resources, Tobacco Research InstituteChinese Academy of Agricultural SciencesQingdaoChina
| | - Cun Guo
- Key Laboratory for Tobacco Gene Resources, Tobacco Research InstituteChinese Academy of Agricultural SciencesQingdaoChina
| | - Jia Chen
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Biology, Hunan UniversityChangshaChina
| | - Wenxuan Pu
- Technology CenterChina Tobacco Hunan Industrial Co., Ltd.ChangshaChina
| | - Feng Yu
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Biology, Hunan UniversityChangshaChina
- Yuelushan LaboratoryChangshaChina
| |
Collapse
|
24
|
Recent Advancement in Drug Design and Discovery of Pyrazole Biomolecules as Cancer and Inflammation Therapeutics. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248708. [PMID: 36557840 PMCID: PMC9780894 DOI: 10.3390/molecules27248708] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 11/27/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022]
Abstract
Pyrazole, an important pharmacophore and a privileged scaffold of immense significance, is a five-membered heterocyclic moiety with an extensive therapeutic profile, viz., anti-inflammatory, anti-microbial, anti-anxiety, anticancer, analgesic, antipyretic, etc. Due to the expansion of pyrazolecent red pharmacological molecules at a quicker pace, there is an urgent need to put emphasis on recent literature with hitherto available information to recognize the status of this scaffold for pharmaceutical research. The reported potential pyrazole-containing compounds are highlighted in the manuscript for the treatment of cancer and inflammation, and the results are mentioned in % inhibition of inflammation, % growth inhibition, IC50, etc. Pyrazole is an important heterocyclic moiety with a strong pharmacological profile, which may act as an important pharmacophore for the drug discovery process. In the struggle to cultivate suitable anti-inflammatory and anticancer agents, chemists have now focused on pyrazole biomolecules. This review conceals the recent expansion of pyrazole biomolecules as anti-inflammatory and anticancer agents with an aim to provide better correlation among different research going around the world.
Collapse
|
25
|
Fatykhov RF, Khalymbadzha IA, Sharapov AD, Potapova AP, Mochulskaya NN, Tsmokalyuk AN, Ivoilova AV, Mozharovskaia PN, Santra S, Chupakhin ON. MnO 2-Mediated Oxidative Cyclization of "Formal" Schiff's Bases: Easy Access to Diverse Naphthofuro-Annulated Triazines. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27207105. [PMID: 36296698 PMCID: PMC9611995 DOI: 10.3390/molecules27207105] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022]
Abstract
A different type of MnO2-induced oxidative cyclization of dihydrotriazines has been developed. These dihydrotriazines are considered as a "formal" Schiff's base. This method provided easy access to naphthofuro-fused triazine via the C-C/C-O oxidative coupling reaction. The reaction sequence comprised the nucleophilic addition of 2-naphthol or phenol to 1,2,4-triazine, followed by oxidative cyclization. The scope and limitations of this novel coupling reaction have been investigated. Further application of the synthesized compound has been demonstrated by synthesizing carbazole-substituted benzofuro-fused triazines. The scalability of the reaction was demonstrated at a 40 mmol load. The mechanistic study strongly suggests that this reaction proceeds through the formation of an O-coordinated manganese complex.
Collapse
Affiliation(s)
- Ramil F. Fatykhov
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University, 19 Mira Str., 620002 Ekaterinburg, Russia
| | - Igor A. Khalymbadzha
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University, 19 Mira Str., 620002 Ekaterinburg, Russia
- Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22 S. Kovalevskaya Str., 620990 Ekaterinburg, Russia
| | - Ainur D. Sharapov
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University, 19 Mira Str., 620002 Ekaterinburg, Russia
| | - Anastasia P. Potapova
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University, 19 Mira Str., 620002 Ekaterinburg, Russia
| | - Nataliya N. Mochulskaya
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University, 19 Mira Str., 620002 Ekaterinburg, Russia
| | - Anton N. Tsmokalyuk
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University, 19 Mira Str., 620002 Ekaterinburg, Russia
| | - Alexandra V. Ivoilova
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University, 19 Mira Str., 620002 Ekaterinburg, Russia
| | - Polina N. Mozharovskaia
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University, 19 Mira Str., 620002 Ekaterinburg, Russia
| | - Sougata Santra
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University, 19 Mira Str., 620002 Ekaterinburg, Russia
- Correspondence:
| | - Oleg N. Chupakhin
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University, 19 Mira Str., 620002 Ekaterinburg, Russia
- Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22 S. Kovalevskaya Str., 620990 Ekaterinburg, Russia
| |
Collapse
|
26
|
Feng Y, Yang Z, Xu X. c-Met: A Promising Therapeutic Target in Bladder Cancer. Cancer Manag Res 2022; 14:2379-2388. [PMID: 35967753 PMCID: PMC9374328 DOI: 10.2147/cmar.s369175] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal-epithelial transition factor (c-Met) belongs to the tyrosine kinase receptor family and is overexpressed in various human cancers. Its ligand is hepatocyte growth factor (HGF), and the HGF/c-Met signaling pathway is involved in a wide range of cellular processes, including cell proliferation, migration, and metastasis. Emerging studies have indicated that c-Met expression is strongly associated with bladder cancer (BCa) development and prognosis. Therefore, c-Met is a potential therapeutic target for BCa treatment. Recently, the aberrant expression of noncoding RNAs was found to play a significant role in tumour progression. There is a close connection between c-Met and noncoding RNA. Herein, we summarized the biological function and prognostic value of c-Met in BCa, as well as its potential role as a drug target. The relation of c-Met and ncRNA was also described in the paper.
Collapse
Affiliation(s)
- Yanfei Feng
- The Second Affiliated College, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Zitong Yang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Xin Xu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
27
|
Harrold E, Corrigan L, Barry S, Lowery M. Targeting MET amplification in Gastro-oesophageal (GO) malignancies and overcoming MET inhibitor resistance: challenges and opportunities. Expert Rev Gastroenterol Hepatol 2022; 16:601-624. [PMID: 35757852 DOI: 10.1080/17474124.2022.2093185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION MET, the hepatocyte growth factor receptor is amplified in 8% of gastroesophageal (GO) malignancies and associated with poor prognosis. Therapeutic targeting of MET amplification and MET mutations has the potential to improve outcomes for patients with GO cancers (GOC). AREAS COVERED The efficacy of MET inhibition (METi) in preclinical studies has yet to translate into meaningful improvements in the treatment paradigm for unselected GOC. MET amplification has been proposed as a superior modality for patient selection; however even if confirmed, frequency and duration of response to METi are limited by rapid activation of primary and secondary resistance pathways. These observations illustrate the challenges inherent in the application of precision oncology predicated on the theory of oncogenic addiction. EXPERT OPINION A standardized definition of MET positivity is critical to enhance patient selection. Early successes targeting the METex14 skipping mutation demonstrate the potent therapeutic effects of METi in a clearly molecularly defined cohort. There is robust preclinical rationale and early-phase data supporting exploitation of immune system interaction with MET. Pragmatic investigation of rational therapeutic combinations based on molecular profiling of both primary and metastatic disease sites with sequential circulating tumor DNA analysis can inform successful clinical development of METi agents in GOC.
Collapse
Affiliation(s)
- Emily Harrold
- Medical Oncology Department, Mater Private Hospital Dublin, Leinster, Ireland.,Trinity St James Cancer Institute, Trinity College Dublin, Leinster, Ireland
| | - Lynda Corrigan
- Trinity St James Cancer Institute, Trinity College Dublin, Leinster, Ireland.,Medical Oncology Department, Tallaght/AMNCH Hospital Dublin, Leinster, Ireland
| | - Simon Barry
- Medical Oncology Department, St James University Hospital Dublin, Leinster, Ireland
| | - Maeve Lowery
- Trinity St James Cancer Institute, Trinity College Dublin, Leinster, Ireland.,Medical Oncology Department, St James University Hospital Dublin, Leinster, Ireland
| |
Collapse
|
28
|
Qi B, Wang F, He H, Fan M, Hu L, Xiong L, Gong G, Shi S, Song X. Identification of (S)-1-(2-(2,4-difluorophenyl)-4-oxothiazolidin-3-yl)-3-(4-((7-(3-(4-ethylpiperazin-1-yl)propoxy)-6-methoxyquinolin-4-yl)oxy)-3,5-difluorophenyl)urea as a potential anti-colorectal cancer agent. Eur J Med Chem 2022; 239:114561. [PMID: 35763868 DOI: 10.1016/j.ejmech.2022.114561] [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: 04/18/2022] [Revised: 06/12/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
In our previous study, 1-(2-(2,6-difluorophenyl)-4-oxothiazolidin-3-yl)-3-(4-((7-(3-(4-ethylpiperazin-1-yl)propoxy)-6-methoxyquinolin-4-yl)oxy)-3,5-difluorophenyl)urea (1) was obtained as a potent tyrosine kinase inhibitor. Further structural optimization was performed in this investigation, and a series of novel quinoline derivates were designed, synthesized and evaluated for their biological activity. Among them, compound 8m possessed nanomolar c-Met and Ron inhibitory activity, with IC50 values of 4.32 nM and 2.39 nM, respectively. Kinase profile study demonstrated that it could also inhibit ABL, PDGFRβ, AXL, RET, and FLT3 with submicromolar potency. It also exhibited moderate to excellent cytotoxic activity against different types of human cancer cell lines, especially against COLO 205 cells (IC50 = 0.035 μM) which was remarkably superior to that of Cabozantinib (IC50 = 6.6 μM) and Fruquintinib (IC50 > 10.0 μM). Compared to ( ± )-8m, isomer (S)-8m and (R)-8m showed similar kinase inhibitory activity against c-Met/RON and in vitro anticancer activity against COLO 205 cells. Differently, compound (S)-8m showed an over 238-fold selectivity toward COLO 205 (IC50 = 0.042 μM) cells to FHC cells (IC50 > 10.0 μM), which indicated its low cytotoxicity against human normal tissue cells. Flow cytometry study demonstrated that compound (S)-8m could significantly induce apoptosis in COLO 205 cells in a dose-dependent manner. Cell cycle arrest assays showed that compound (S)-8m could not arrest the cell-cycle progression due to the massive dead cells.
Collapse
Affiliation(s)
- Baohui Qi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China.
| | - Fei Wang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Huan He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Mengmeng Fan
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Liping Hu
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Li Xiong
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Guowei Gong
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Shengmin Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Xiaomeng Song
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| |
Collapse
|
29
|
Fujino T, Suda K, Koga T, Hamada A, Ohara S, Chiba M, Shimoji M, Takemoto T, Soh J, Mitsudomi T. Foretinib can overcome common on-target resistance mutations after capmatinib/tepotinib treatment in NSCLCs with MET exon 14 skipping mutation. J Hematol Oncol 2022; 15:79. [PMID: 35690785 PMCID: PMC9188708 DOI: 10.1186/s13045-022-01299-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/01/2022] [Indexed: 02/04/2023] Open
Abstract
Background Capmatinib and tepotinib are guideline-recommended front-line treatments for non-small-cell lung cancer (NSCLC) patients with MET exon 14 skipping mutations (METex14). However, the emergence of acquired resistance to capmatinib/tepotinib is almost inevitable partially due to D1228X or Y1230X secondary mutations of the MET. In this study, we explored agents that are active against both D1228X and Y1230X MET to propose an ideal sequential treatment after capmatinib/tepotinib treatment failure in NSCLC patients with METex14. Methods The inhibitory effects of 300 drugs, including 33 MET-TKIs, were screened in Ba/F3 cells carrying METex14 plus MET D1228A/Y secondary mutations. The screen revealed four-candidate type II MET-TKIs (altiratinib, CEP-40783, foretinib and sitravatinib). Therefore, we performed further growth inhibitory assays using these four MET-TKIs plus cabozantinib and merestinib in Ba/F3 cells carrying MET D1228A/E/G/H/N/V/Y or Y1230C/D/H/N/S secondary mutations. We also performed analyses using Hs746t cell models carrying METex14 (with mutant allele amplification) with/without D1228X or Y1230X in vitro and in vivo to confirm the findings. Furthermore, molecular dynamics (MD) simulations were carried out to examine differences in binding between type II MET-TKIs. Results All 6 type II MET-TKIs were active against Y1230X secondary mutations. However, among these 6 agents, only foretinib showed potent activity against D1228X secondary mutations of the MET in the Ba/F3 cell and Hs746t in vitro model and Hs746t in vivo model, and CEP-40783 and altiratinib demonstrated some activity. MD analysis suggested that the long tail of foretinib plays an important role in binding D1228X MET through interaction with a residue at the solvent front (G1163). Tertiary G1163X mutations, together with L1195F/I and F1200I/L, occurred as acquired resistance mechanisms to the second-line treatment foretinib in Ba/F3 cell models. Conclusions The type II MET-TKI foretinib may be an appropriate second-line treatment for NSCLCs carrying METex14 after campatinib/tepotinib treatment failure by secondary mutations at residue D1228 or Y1230. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01299-z.
Collapse
Affiliation(s)
- Toshio Fujino
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Kenichi Suda
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Takamasa Koga
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Akira Hamada
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Shuta Ohara
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Masato Chiba
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Masaki Shimoji
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Toshiki Takemoto
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Junichi Soh
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Tetsuya Mitsudomi
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan.
| |
Collapse
|
30
|
GEINDREAU M, BRUCHARD M, VEGRAN F. Role of Cytokines and Chemokines in Angiogenesis in a Tumor Context. Cancers (Basel) 2022; 14:cancers14102446. [PMID: 35626056 PMCID: PMC9139472 DOI: 10.3390/cancers14102446] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Tumor growth in solid cancers requires adequate nutrient and oxygen supply, provided by blood vessels created by angiogenesis. Numerous studies have demonstrated that this mechanism plays a crucial role in cancer development and appears to be a well-defined hallmark of cancer. This process is carefully regulated, notably by cytokines with pro-angiogenic or anti-angiogenic features. In this review, we will discuss the role of cytokines in the modulation of angiogenesis. In addition, we will summarize the therapeutic approaches based on cytokine modulation and their clinical approval. Abstract During carcinogenesis, tumors set various mechanisms to help support their development. Angiogenesis is a crucial process for cancer development as it drives the creation of blood vessels within the tumor. These newly formed blood vessels insure the supply of oxygen and nutrients to the tumor, helping its growth. The main factors that regulate angiogenesis are the five members of the vascular endothelial growth factor (VEGF) family. Angiogenesis is a hallmark of cancer and has been the target of new therapies this past few years. However, angiogenesis is a complex phenomenon with many redundancy pathways that ensure its maintenance. In this review, we will first describe the consecutive steps forming angiogenesis, as well as its classical regulators. We will then discuss how the cytokines and chemokines present in the tumor microenvironment can induce or block angiogenesis. Finally, we will focus on the therapeutic arsenal targeting angiogenesis in cancer and the challenges they have to overcome.
Collapse
Affiliation(s)
- Mannon GEINDREAU
- Université de Bourgogne Franche-Comté, 21000 Dijon, France; (M.G.); (M.B.)
- CRI INSERM UMR1231 ‘Lipids, Nutrition and Cancer’ Team CAdiR, 21000 Dijon, France
| | - Mélanie BRUCHARD
- Université de Bourgogne Franche-Comté, 21000 Dijon, France; (M.G.); (M.B.)
- CRI INSERM UMR1231 ‘Lipids, Nutrition and Cancer’ Team CAdiR, 21000 Dijon, France
- Centre Georges-François Leclerc, UNICANCER, 21000 Dijon, France
- LipSTIC Labex, 21000 Dijon, France
| | - Frédérique VEGRAN
- Université de Bourgogne Franche-Comté, 21000 Dijon, France; (M.G.); (M.B.)
- CRI INSERM UMR1231 ‘Lipids, Nutrition and Cancer’ Team CAdiR, 21000 Dijon, France
- Centre Georges-François Leclerc, UNICANCER, 21000 Dijon, France
- LipSTIC Labex, 21000 Dijon, France
- Correspondence:
| |
Collapse
|
31
|
Gu W, Shen H, Xie L, Zhang X, Yang J. The Role of Feedback Loops in Targeted Therapy for Pancreatic Cancer. Front Oncol 2022; 12:800140. [PMID: 35651786 PMCID: PMC9148955 DOI: 10.3389/fonc.2022.800140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic cancer is the leading cause of cancer-related deaths worldwide, with limited treatment options and low long-term survival rates. The complex and variable signal regulation networks are one of the important reasons why it is difficult for pancreatic cancer to develop precise targeted therapy drugs. Numerous studies have associated feedback loop regulation with the development and therapeutic response of cancers including pancreatic cancer. Therefore, we review researches on the role of feedback loops in the progression of pancreatic cancer, and summarize the connection between feedback loops and several signaling pathways in pancreatic cancer, as well as recent advances in the intervention of feedback loops in pancreatic cancer treatment, highlighting the potential of capitalizing on feedback loops modulation in targeted therapy for pancreatic cancer.
Collapse
Affiliation(s)
- Weigang Gu
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - HongZhang Shen
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lu Xie
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- *Correspondence: Xiaofeng Zhang, ; Jianfeng Yang,
| | - Jianfeng Yang
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Xiaofeng Zhang, ; Jianfeng Yang,
| |
Collapse
|
32
|
Zhao Y, Ye W, Wang YD, Chen WD. HGF/c-Met: A Key Promoter in Liver Regeneration. Front Pharmacol 2022; 13:808855. [PMID: 35370682 PMCID: PMC8968572 DOI: 10.3389/fphar.2022.808855] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/11/2022] [Indexed: 01/18/2023] Open
Abstract
Hepatocyte growth factor (HGF) is a peptide-containing multifunctional cytokine that acts on various epithelial cells to regulate cell growth, movement and morphogenesis, and tissue regeneration of injured organs. HGF is sequestered by heparin-like protein in its inactive form and is widespread in the extracellular matrix of most tissues. When the liver loses its average mass, volume, or physiological and biochemical functions due to various reasons, HGF binds to its specific receptor c-Met (cellular mesenchymal-epithelial transition) and transmits the signals into the cells, and triggers the intrinsic kinase activity of c-Met. The downstream cascades of HGF/c-Met include JAK/STAT3, PI3K/Akt/NF-κB, and Ras/Raf pathways, affecting cell proliferation, growth, and survival. HGF has important clinical significance for liver fibrosis, hepatocyte regeneration after inflammation, and liver regeneration after transplantation. And the development of HGF as a biological drug for regenerative therapy of diseases, that is, using recombinant human HGF protein to treat disorders in clinical trials, is underway. This review summarizes the recent findings of the HGF/c-Met signaling functions in liver regeneration.
Collapse
Affiliation(s)
- Yang Zhao
- Key Laboratory of Receptors-Mediated Gene Regulation, The People's Hospital of Hebi, School of Medicine, Henan University, Kaifeng, China
| | - Wenling Ye
- Key Laboratory of Receptors-Mediated Gene Regulation, The People's Hospital of Hebi, School of Medicine, Henan University, Kaifeng, China
| | - Yan-Dong Wang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Wei-Dong Chen
- Key Laboratory of Receptors-Mediated Gene Regulation, The People's Hospital of Hebi, School of Medicine, Henan University, Kaifeng, China
| |
Collapse
|
33
|
Design, synthesis and anticancer evaluation of 6,7-disubstituted-4-phenoxyquinoline derivatives bearing 1,8-naphthyridine-3-carboxamide moiety as novel multi-target TKIs. Bioorg Chem 2022; 121:105672. [PMID: 35202851 DOI: 10.1016/j.bioorg.2022.105672] [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: 10/14/2021] [Revised: 01/01/2022] [Accepted: 02/07/2022] [Indexed: 12/20/2022]
Abstract
Giving the fact that the disorders of multiple receptor tyrosine kinases (RTKs) are characteristics of various cancers, we assumed that developing novel multi-target drugs might have an advantage in treating the complex cancers. Taking the multi-target c-Met inhibitor Foretinib as the leading compound, we discovered a novel series of 6,7-disubstituted-4-phenoxyquinoline derivatives bearing 1,8-naphthyridine-3-carboxamide moiety with the help of molecular docking. Among them, the most promising compound 33 showed a prominent activity against Hela (IC50 = 0.21 µM), A549 (IC50 = 0.39 µM), and MCF-7 (IC50 = 0.33 µM), which were 3.28-4.82 times more active than that of Foretinib. Additionally, compound 33 dose dependently induced apoptosis by arresting A549 cells at G1 phase. Enzymatic assays and docking analyses were further confirmed that compound 33 was a multi-target inhibitor with the strong potencies against c-Met (IC50 = 11.77 nM), MEK1 (IC50 = 10.71 nM), and Flt-3 (IC50 = 22.36 nM). In the A549 cells mediated xenograft mouse model, compound 33 inhibited the tumor growth (TGI = 64%) without obvious toxicity, establishing compound 33 as a promising candidate for cancer therapy.
Collapse
|
34
|
Liang D, Yu C, Qin X, Yang X, Dong X, Hu M, Du L, Li M. Discovery of small-molecule fluorescent probes for C-Met. Eur J Med Chem 2022; 230:114114. [PMID: 35051746 DOI: 10.1016/j.ejmech.2022.114114] [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: 06/16/2021] [Revised: 12/23/2021] [Accepted: 01/09/2022] [Indexed: 11/17/2022]
Abstract
C-mesenchymal-epithelia transition factor (c-Met) is highly expressed in various solid tumors such as gastric cancer, liver cancer, and lung cancer, playing a pivotal role in the growth, maintenance, and development of different tumor cells. In this study, three small-molecule fluorescent probes (5, 11, 16) targeting c-Met were developed, and their design strategies were also initially explored. In general, the fluorescence properties of the probes themselves could meet the imaging requirements, and they have shown sufficient inhibitory activities against c-Met, especially probe 16, reflecting the targeting and acceptance. Also, fluorescence polarization assays and flow cytometry analysis verified the binding between the probes and c-Met. Cell imaging confirmed that these probes could be used to label c-Met on living cells. It is of positive significance for the development of c-Met kinase inhibitors and tumor pathology research.
Collapse
Affiliation(s)
- Dong Liang
- Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Chen Yu
- Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaojun Qin
- Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xingye Yang
- Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xuhui Dong
- Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mingzhao Hu
- Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lupei Du
- Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Minyong Li
- Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
35
|
Ma Q, Kong L, Zhong D. Case Report: Dramatic Response to Crizotinib in a Patient With Non-Small Cell Lung Cancer Positive for a Novel ARL1-MET Fusion. Front Oncol 2022; 12:804330. [PMID: 35237515 PMCID: PMC8883050 DOI: 10.3389/fonc.2022.804330] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
It is imperative to know the status of oncogenic drivers in patients with non-small cell lung cancer (NSCLC). Compared with ALK and ROS1 fusion, MET fusion is relatively rare in NSCLC. In this case, we report the case of a female patient with NSCLC positive for a novel ARL1-MET fusion. The patient achieved about a 5-month progression-free survival (PFS) after receiving crizotinib for unresectable right lung malignancies. To the best of our knowledge, this case provides the first clinical evidence that the novel ARL1-MET fusion might be an actionable mutation in NSCLC.
Collapse
|
36
|
Wade Wolfe MM, Guo S, Yu LS, Vogel TR, Tucker JW, Szymczak NK. Nucleophilic strategies to construct –CF 2– linkages using borazine-CF 2Ar reagents. Chem Commun (Camb) 2022; 58:11705-11708. [DOI: 10.1039/d2cc01938h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Using nucleophilic, boron-based –CF2Ar reagents, we demonstrate three methods to form C–CF bonds: (1) nucleophilic aromatic substitution, (2) palladium catalyzed cross-coupling, and (3) nucleophilic substitution.
Collapse
Affiliation(s)
| | - Shuo Guo
- University of Michigan, 930 N. University Ave., Ann Arbor, MI, 48109, USA
| | - Lucy S. Yu
- University of Michigan, 930 N. University Ave., Ann Arbor, MI, 48109, USA
| | - Trenton R. Vogel
- University of Michigan, 930 N. University Ave., Ann Arbor, MI, 48109, USA
| | - Joseph W. Tucker
- Medicine Design, Pfizer Inc., Eastern Point Rd., Groton, CT, 06340, USA
| | | |
Collapse
|
37
|
Batra U, Nathany S. MET: A narrative review of exon 14 skipping mutation in non-small-cell lung carcinoma. CANCER RESEARCH, STATISTICS, AND TREATMENT 2022. [DOI: 10.4103/crst.crst_158_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
38
|
Huang X, Guo J, Li T, Jia L, Tang X, Zhu J, Tang Q, Feng Z. c-Met-targeted chimeric antigen receptor T cells inhibit hepatocellular carcinoma cells in vitro and in vivo. J Biomed Res 2021; 36:10-21. [PMID: 35403606 PMCID: PMC8894281 DOI: 10.7555/jbr.35.20200207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
c-Met is a hepatocyte growth factor receptor overexpressed in many tumors such as hepatocellular carcinoma (HCC). Therefore, c-Met may serve as a promising target for HCC immunotherapy. Modifying T cells to express c-Met-specific chimeric antigen receptor (CAR) is an attractive strategy in treating c-Met-positive HCC. This study aimed to systematically evaluate the inhibitory effects of 2nd- and 3rd-generation c-Met CAR-T cells on hepatocellular carcinoma (HCC) cells. Here, 2nd- and 3rd-generation c-Met CARs containing an anti-c-Met single-chain variable fragment (scFv) as well as the CD28 signaling domain and CD3ζ (c-Met-28-3ζ), the CD137 signaling domain and CD3ζ (c-Met-137-3ζ), or the CD28 and CD137 signaling domains and CD3ζ (c-Met-28-137-3ζ) were constructed, and their abilities to target c-Met-positive HCC cells were evaluated in vitro and in vivo. All c-Met CARs were stably expressed on T cell membrane, and c-Met CAR-T cells aggregated around c-Met-positive HCC cells and specifically killed them in vitro. c-Met-28-137-3ζ CAR-T cells secreted more interferon-gamma (IFN-γ) and interleukin 2 (IL-2) than c-Met-28-3ζ CAR-T cells and c-Met-137-3ζ CAR-T cells. Compared with c-Met low-expressed cells, c-Met CAR-T cells secreted more cytokines when co-cultured with c-Met high-expressed cells. Moreover, c-Met-28-137-3ζ CAR-T cells eradicated HCC more effectively in xenograft tumor models compared with the control groups. This study suggests that 3rd-generation c-Met CAR-T cells are more effective in inhibiting c-Met-positive HCC cells than 2nd-generation c-Met CAR-T cells, thereby providing a promising therapeutic intervention for c-Met-positive HCC.
Collapse
Affiliation(s)
- Xiaochen Huang
- National Health Commission Key Laboratory of Antibody Techniques, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Departments of Pathology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Jiangsu Province Institute of Cancer, Nanjing, Jiangsu 210009, China
| | - Jiaojiao Guo
- National Health Commission Key Laboratory of Antibody Techniques, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Tao Li
- National Health Commission Key Laboratory of Antibody Techniques, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Lizhou Jia
- National Health Commission Key Laboratory of Antibody Techniques, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaojun Tang
- National Health Commission Key Laboratory of Antibody Techniques, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jin Zhu
- Huadong Medical Institute of Biotechniques, Nanjing, Jiangsu 210002, China
| | - Qi Tang
- National Health Commission Key Laboratory of Antibody Techniques, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zhenqing Feng
- National Health Commission Key Laboratory of Antibody Techniques, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
39
|
Craig SG, Mende S, Humphries MP, Bingham V, Viratham Pulsawatdi A, Loughrey MB, Coleman HG, McQuaid S, Wilson RH, Van Schaeybroeck S, James JA, Salto‐Tellez M. Orthogonal MET analysis in a population-representative stage II-III colon cancer cohort: prognostic and potential therapeutic implications. Mol Oncol 2021; 15:3317-3328. [PMID: 34428346 PMCID: PMC8637556 DOI: 10.1002/1878-0261.13089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/23/2021] [Indexed: 01/11/2023] Open
Abstract
Clinical trials for MET inhibitors have demonstrated limited success for their use in colon cancer (CC). However, clinical efficacy may be obscured by a lack of standardisation in MET assessment for patient stratification. In this study, we aimed to determine the molecular context in which MET is deregulated in CC using a series of genomic and proteomic tests to define MET expression and identify patient subgroups that should be considered in future studies with MET-targeted agents. To this aim, orthogonal expression analysis of MET was conducted in a population-representative cohort of stage II/III CC patients (n = 240) diagnosed in Northern Ireland from 2004 to 2008. Targeted sequencing was used to determine the relative incidence of MET R970C and MET T992I mutations within the cohort. MET amplification was assessed using dual-colour dual-hapten brightfield in situ hybridisation (DDISH). Expression of transcribed MET and c-MET protein within the cohort was assessed using digital image analysis on MET RNA in situ hybridisation (ISH) and c-MET immunohistochemistry (IHC) stained slides. We found that less than 2% of the stage II/III CC patient population assessed demonstrated a genetic MET aberration. Determination of a high MET RNA-ISH/low c-MET IHC protein subgroup was found to be associated with poor 5-year cancer-specific outcomes compared to patients with concordant MET RNA-ISH and c-MET IHC protein expression (HR 2.12 [95%CI: 1.27-3.68]). The MET RNA-ISH/c-MET IHC protein biomarker paradigm identified in this study demonstrates that subtyping of MET expression may be required to identify MET-addicted malignancies in CC patients who will truly benefit from MET inhibition.
Collapse
Affiliation(s)
- Stephanie G. Craig
- Precision Medicine Centre of ExcellencePatrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Svenja Mende
- Precision Medicine Centre of ExcellencePatrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Matthew P. Humphries
- Precision Medicine Centre of ExcellencePatrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Victoria Bingham
- Precision Medicine Centre of ExcellencePatrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Amélie Viratham Pulsawatdi
- Precision Medicine Centre of ExcellencePatrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Maurice B. Loughrey
- Department of Cellular PathologyRoyal Victoria HospitalBelfast Health and Social Care TrustBelfastUK
- Centre for Public HealthQueen's University BelfastUK
| | | | - Stephen McQuaid
- Precision Medicine Centre of ExcellencePatrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | | | - Sandra Van Schaeybroeck
- Precision Medicine Centre of ExcellencePatrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Jacqueline A. James
- Precision Medicine Centre of ExcellencePatrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
- Department of Cellular PathologyRoyal Victoria HospitalBelfast Health and Social Care TrustBelfastUK
| | - Manuel Salto‐Tellez
- Precision Medicine Centre of ExcellencePatrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
- Department of Cellular PathologyRoyal Victoria HospitalBelfast Health and Social Care TrustBelfastUK
| |
Collapse
|
40
|
Bahcall M, Paweletz CP, Kuang Y, Taus LJ, Sim T, Kim ND, Dholakia KH, Lau CJ, Gokhale PC, Chopade PR, Hong F, Wei Z, Köhler J, Kirschmeier PT, Guo J, Guo S, Wang S, Janne PA. Combination of type I and type II MET tyrosine kinase inhibitors as therapeutic approach to prevent resistance. Mol Cancer Ther 2021; 21:322-335. [PMID: 34789563 DOI: 10.1158/1535-7163.mct-21-0344] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 10/13/2021] [Accepted: 11/08/2021] [Indexed: 11/16/2022]
Abstract
MET targeted therapies are clinically effective in MET amplified and MET exon 14 deletion mutant (METex14) non-small cell lung cancers (NSCLC) but their efficacy is limited by the development of drug resistance. Structurally distinct MET tyrosine kinase inhibitors (TKIs) (type I/II) have been developed or are under clinical evaluation, which may overcome MET mediated drug resistance mechanisms. In this study, we assess secondary MET mutations likely to emerge in response to treatment with single-agent or combinations of type I/type II MET TKIs using TPR-MET transformed Ba/F3 cell mutagenesis assays. We found that these inhibitors gave rise to distinct secondary MET mutant profiles. However, a combination of type I/II TKI inhibitors (capmatinib and merestinib) yielded no resistant clones in vitro. The combination of capmatinib/merestinib was evaluated in vivo and led to a significant reduction in tumor outgrowth compared to either MET inhibitor alone. Our findings demonstrate in vitro and in vivo that a simultaneous treatment with a type I and type II MET TKI may be a clinically viable approach to delay and/or diminish the emergence of on target MET mediated drug resistance mutations.
Collapse
Affiliation(s)
- Magda Bahcall
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute
| | - Cloud P Paweletz
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute and Harvard Medical School
| | - Yanan Kuang
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute
| | - Luke J Taus
- Medical Oncology, Dana-Farber Cancer Institute
| | - Taebo Sim
- Severance Biomedical Science Institute, Yonsei University College of Medicine
| | - Nam Doo Kim
- Daegu-Gyeongbuk Medical Innovation Foundation
| | | | - Christie J Lau
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute
| | | | - Pratik R Chopade
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute
| | | | - Zihan Wei
- Biostatistics, Dana-Farber Cancer Institute
| | - Jens Köhler
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | | | | | - Sujuan Guo
- Fralin Biomedical Research Institute, Virginia Tech
| | - Stephen Wang
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute and Harvard Medical School
| | - Pasi A Janne
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute
| |
Collapse
|
41
|
Karmacharya U, Guragain D, Chaudhary P, Jee JG, Kim JA, Jeong BS. Novel Pyridine Bioisostere of Cabozantinib as a Potent c-Met Kinase Inhibitor: Synthesis and Anti-Tumor Activity against Hepatocellular Carcinoma. Int J Mol Sci 2021; 22:ijms22189685. [PMID: 34575841 PMCID: PMC8468607 DOI: 10.3390/ijms22189685] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/22/2022] Open
Abstract
Two novel bioisosteres of cabozantinib, 3 and 4, were designed and synthesized. The benzene ring in the center of the cabozantinib structure was replaced by trimethylpyridine (3) and pyridine (4), respectively. Surprisingly, the two compounds showed extremely contrasting mesenchymal-epithelial transition factor (c-Met) inhibitory activities at 1 μM concentration (4% inhibition of 3 vs. 94% inhibition of 4). The IC50 value of compound 4 was 4.9 nM, similar to that of cabozantinib (5.4 nM). A ligand-based docking study suggested that 4 includes the preferred conformation for the binding to c-Met in the conformational ensemble, but 3 does not. The anti-proliferative activity of compound 4 against hepatocellular carcinoma (Hep3B and Huh7) and non-small-cell lung cancer (A549 and H1299) cell lines was better than that of cabozantinib, whereas 3 did not show a significant anti-proliferative activity. Moreover, the tumor selectivity of compound 4 toward hepatocellular carcinoma cell lines was higher than that of cabozantinib. In the xenograft chick tumor model, compound 4 inhibited Hep3B tumor growth to a much greater extent than cabozantinib. The present study suggests that compound 4 may be a good therapeutic candidate against hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ujjwala Karmacharya
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Korea; (U.K.); (D.G.); (P.C.)
| | - Diwakar Guragain
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Korea; (U.K.); (D.G.); (P.C.)
| | - Prakash Chaudhary
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Korea; (U.K.); (D.G.); (P.C.)
| | - Jun-Goo Jee
- College of Pharmacy, Kyungpook National University, Daegu 41566, Korea;
| | - Jung-Ae Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Korea; (U.K.); (D.G.); (P.C.)
- Correspondence: (J.-A.K.); (B.-S.J.); Tel.: +82-53-810-2816 (J.-A.K.); +82-53-810-2814 (B.-S.J.)
| | - Byeong-Seon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Korea; (U.K.); (D.G.); (P.C.)
- Correspondence: (J.-A.K.); (B.-S.J.); Tel.: +82-53-810-2816 (J.-A.K.); +82-53-810-2814 (B.-S.J.)
| |
Collapse
|
42
|
Xu C, Liu Y, Zhao G. The development of 3-substituted indolin-2-one derivatives as kinase inhibitors for cancer therapy. Curr Med Chem 2021; 29:1891-1919. [PMID: 34465277 DOI: 10.2174/0929867328666210831142311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 11/22/2022]
Abstract
Kinases are pivotal regulators in tumorigenesis and metastasis by modulating the expression of oncogenes and the transcription of antioncogenes directly or indirectly. Correspondingly, multifarious 3-substituted indolin-2-one derivatives as selective kinase inhibitors for cancer therapy exhibited a low nanomolar activity with prominent efficacy, superior response rate and admirable tolerability. Particularly, certain 3-substituted indolin-2-one derivatives have met the requirements for clinical trials or the pharmaceutical market. Herein, we focus on the traits of 3-substituted indolin-2-one derivatives as kinase inhibitors for cancer therapy, overview recent progress of 3-substituted indolin-2-one derivatives as kinase inhibitors for cancer therapy, analyze the selectivity for tyrosine kinases inhibitors and serine/threonine kinases inhibitors from the molecular aspects based on the molecular docking studies, summarize the structure-activity relationships (SARs) as selective kinase inhibitors and provide our perspectives for the development of 3-substituted indolin-2-one derivatives as kinase inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Changqing Xu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong. China
| | - Yang Liu
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas. United States
| | - Guisen Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong. China
| |
Collapse
|
43
|
Design, Synthesis and Biological Evaluation of Novel
α‐Acyloxycarboxamide‐Based
Derivatives as
c‐Met
Inhibitors. CHINESE J CHEM 2021. [DOI: 10.1002/cjoc.202100106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
44
|
Shi T, Hu J, Wang W, Jiang Q, Xu Z, Yu S, Wang F, Liu X. Multiple Blockades of the HGF/Met Signaling Pathway for Metastasis Suppression Using Nanoinhibitors. ACS APPLIED MATERIALS & INTERFACES 2021; 13:30350-30358. [PMID: 34165951 DOI: 10.1021/acsami.1c07010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The hepatocyte growth factor (HGF)/HGF receptor (Met) signaling pathway serves as a potential target for preventing tumor metastasis yet poorly explored. Here, we developed a Met-targeted nanoinhibitor to efficiently suppress metastasis via a multiple blockading HGF/Met signaling pathway. A biocompatible nanovector comprising multiple type of inhibitors enables interrupting extracellular domain dimerization and intracellular domain phosphorylation simultaneously. Such a comprehensive blockade of signaling pathway restrains unregulated tumor cell migration, invasion, and proliferation and thus remarkably suppresses metastasis in an orthotopic breast tumor model. This method provides a safe and effective option for metastasis inhibition via modulation of the cell signaling pathway. To our best knowledge, the strategy of the multiple blockading signaling pathway has not been reported for preventing tumor metastasis.
Collapse
Affiliation(s)
- Tianhui Shi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Jialing Hu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Wenxiao Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Qunying Jiang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Zhen Xu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Shuyi Yu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Xiaoqing Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
45
|
Aftimos P, Rolfo C, Rottey S, Barthélémy P, Borg C, Park K, Oh DY, Kim SW, De Jonge N, Hanssens V, Zwanenpoel K, Molthoff C, Vugts D, Dreier T, Verheesen P, van Dongen GA, Jacobs J, Van Rompaey L, Hultberg A, Michieli P, Pauwels P, Fung S, Thibault A, de Haard H, Leupin N, Awada A. The NHance ® Mutation-Equipped Anti-MET Antibody ARGX-111 Displays Increased Tissue Penetration and Anti-Tumor Activity in Advanced Cancer Patients. Biomedicines 2021; 9:biomedicines9060665. [PMID: 34200749 PMCID: PMC8229762 DOI: 10.3390/biomedicines9060665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/27/2021] [Accepted: 06/08/2021] [Indexed: 11/25/2022] Open
Abstract
Dysregulation of MET signaling has been implicated in tumorigenesis and metastasis. ARGX-111 combines complete blockade of this pathway with enhanced tumor cell killing and was investigated in 24 patients with MET-positive advanced cancers in a phase 1b study at four dose levels (0.3–10 mg/kg). ARGX-111 was well tolerated up to 3 mg/kg (MTD). Anti-tumor activity was observed in nearly half of the patients (46%) with a mean duration of treatment of 12 weeks. NHance® mutations in the Fc of ARGX-111 increased affinity for the neonatal Fc receptor (FcRn) at acidic pH, stimulating transcytosis across FcRn-expressing cells and radiolabeled ARGX-111 accumulated in lymphoid tissues, bone and liver, organs expressing FcRn at high levels in a biodistribution study using human FcRn transgenic mice. In line with this, we observed, in a patient with MET-amplified (>10 copies) gastric cancer, diminished metabolic activity in multiple metastatic lesions in lymphoid and bone tissues by 18F-FDG-PET/CT after two infusions with 0.3 mg/kg ARGX-111. When escalated to 1 mg/kg, a partial response was reached. Furthermore, decreased numbers of CTC (75%) possibly by the enhanced tumor cell killing witnessed the modes of action of the drug, warranting further clinical investigation of ARGX-111.
Collapse
Affiliation(s)
- Philippe Aftimos
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (A.A.)
| | - Christian Rolfo
- University Hospital Antwerp, 2650 Edegem, Belgium; (C.R.); (K.Z.); (P.P.)
| | | | - Philippe Barthélémy
- Medical Oncology Unit, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France;
| | - Christophe Borg
- Medical Oncology Department, University Hospital of Besançon, CEDEX, 25000 Besançon, France;
| | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Do-Youn Oh
- Seoul National University Hospital, Seoul 03080, Korea;
| | - Sang-We Kim
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Natalie De Jonge
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Valérie Hanssens
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Karen Zwanenpoel
- University Hospital Antwerp, 2650 Edegem, Belgium; (C.R.); (K.Z.); (P.P.)
| | - Carla Molthoff
- Department of Radiology & Nuclear Medicine, VU University Medical Center Amsterdam, 1012 Amsterdam, The Netherlands; (C.M.); (D.V.); (G.A.M.S.v.D.)
| | - Daniëlle Vugts
- Department of Radiology & Nuclear Medicine, VU University Medical Center Amsterdam, 1012 Amsterdam, The Netherlands; (C.M.); (D.V.); (G.A.M.S.v.D.)
| | - Torsten Dreier
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
- AgomAb Therapeutics NV, 9000 Ghent, Belgium;
| | - Peter Verheesen
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Guus A.M.S. van Dongen
- Department of Radiology & Nuclear Medicine, VU University Medical Center Amsterdam, 1012 Amsterdam, The Netherlands; (C.M.); (D.V.); (G.A.M.S.v.D.)
| | - Julie Jacobs
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Luc Van Rompaey
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Anna Hultberg
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Paolo Michieli
- AgomAb Therapeutics NV, 9000 Ghent, Belgium;
- Department of Oncology, University of Torino Medical School, 10124 Turin, Italy
| | - Patrick Pauwels
- University Hospital Antwerp, 2650 Edegem, Belgium; (C.R.); (K.Z.); (P.P.)
| | - Samson Fung
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Alain Thibault
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Hans de Haard
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Nicolas Leupin
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
- Correspondence: ; Tel.: +41-79-293-18-14
| | - Ahmad Awada
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (A.A.)
| |
Collapse
|
46
|
Tang Q, Aronov AM, Deininger DD, Giroux S, Lauffer DJ, Li P, Liang J, McGinty K, Ronkin S, Swett R, Waal N, Boucher D, Ford PJ, Moody CS. Discovery of Potent, Selective Triazolothiadiazole-Containing c-Met Inhibitors. ACS Med Chem Lett 2021; 12:955-960. [PMID: 34141080 DOI: 10.1021/acsmedchemlett.1c00094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/11/2021] [Indexed: 12/29/2022] Open
Abstract
Herein, we report a novel series of highly potent and selective triazolothiadiazole c-Met inhibitors. Starting with molecule 5, we have applied structure-based drug design principles to identify the triazolothiadiazole ring system. We successfully replaced the metabolically unstable phenolic moiety with a quinoline group. Further optimization around the 5,6 bicyclic moiety led to the identification of 21. Compound 21 suffered from PDE3 selectivity issues and subsequent, structurally informed design led to the discovery of compound 23. Compound 23 has exquisite kinase selectivity, excellent potency, favorable ADME profile, and showed dose-dependent antitumor efficacy in a SNU-5 gastric cancer xenograft model.
Collapse
Affiliation(s)
- Qing Tang
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Alex M. Aronov
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - David D. Deininger
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Simon Giroux
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - David J. Lauffer
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Pan Li
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Jianglin Liang
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Kira McGinty
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Steven Ronkin
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Rebecca Swett
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Nathan Waal
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Diane Boucher
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Pamella J. Ford
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Cameron S. Moody
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| |
Collapse
|
47
|
Shi M, Ma J, Feng M, Liang L, Chen H, Wang T, Xie Z. Novel MET exon 14 skipping analogs characterized in non-small cell lung cancer patients: A case study. Cancer Genet 2021; 256-257:62-67. [PMID: 33905998 DOI: 10.1016/j.cancergen.2021.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/25/2021] [Accepted: 04/08/2021] [Indexed: 11/29/2022]
Abstract
MET exon 14 skipping (METex14) is a validated oncogenic driver in lung cancer and MET tyrosine kinase inhibitors are now available as effective clinical treatments. The majority of known METex14 alterations are typical donor/acceptor splicing or ubiquitination site mutations. Herein, two new METex14 variants were detected in two patients with lung adenocarcinoma by targeted next generation sequencing (NGS). Reverse transcription (RT)-based analysis confirmed that these mutations led to MET exon 14 skipping. Our analysis provided evidence for possible targeted therapy options for patients carrying these MET mutations or similar METex14 analogs.
Collapse
Affiliation(s)
- Minke Shi
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, China
| | - Jing Ma
- Department of Data System, 3D Medicines Inc., Shanghai, China
| | - Meilin Feng
- Department of Data System, 3D Medicines Inc., Shanghai, China
| | - Lei Liang
- Department of Research and Development, 3D Medicines Inc., Shanghai, China
| | - Hongyuan Chen
- Department of Research and Development, 3D Medicines Inc., Shanghai, China
| | - Tao Wang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, China.
| | - Zhenghua Xie
- Department of Research and Development, 3D Medicines Inc., Shanghai, China.
| |
Collapse
|
48
|
Resistance to Molecularly Targeted Therapies in Melanoma. Cancers (Basel) 2021; 13:cancers13051115. [PMID: 33807778 PMCID: PMC7961479 DOI: 10.3390/cancers13051115] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Malignant melanoma is the most aggressive type of skin cancer with invasive growth patterns. In 2021, 106,110 patients are projected to be diagnosed with melanoma, out of which 7180 are expected to die. Traditional methods like surgery, radiation therapy, and chemotherapy are not effective in the treatment of metastatic and advanced melanoma. Recent approaches to treat melanoma have focused on biomarkers that play significant roles in cell growth, proliferation, migration, and survival. Several FDA-approved molecular targeted therapies such as tyrosine kinase inhibitors (TKIs) have been developed against genetic biomarkers whose overexpression is implicated in tumorigenesis. The use of targeted therapies as an alternative or supplement to immunotherapy has revolutionized the management of metastatic melanoma. Although this treatment strategy is more efficacious and less toxic in comparison to traditional therapies, targeted therapies are less effective after prolonged treatment due to acquired resistance caused by mutations and activation of alternative mechanisms in melanoma tumors. Recent studies focus on understanding the mechanisms of acquired resistance to these current therapies. Further research is needed for the development of better approaches to improve prognosis in melanoma patients. In this article, various melanoma biomarkers including BRAF, MEK, RAS, c-KIT, VEGFR, c-MET and PI3K are described, and their potential mechanisms for drug resistance are discussed.
Collapse
|
49
|
Santarpia M, Massafra M, Gebbia V, D’Aquino A, Garipoli C, Altavilla G, Rosell R. A narrative review of MET inhibitors in non-small cell lung cancer with MET exon 14 skipping mutations. Transl Lung Cancer Res 2021; 10:1536-1556. [PMID: 33889528 PMCID: PMC8044480 DOI: 10.21037/tlcr-20-1113] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Treatment of advanced non-small cell lung cancer (NSCLC) has radically improved in the last years due to development and clinical approval of highly effective agents including immune checkpoint inhibitors (ICIs) and oncogene-directed therapies. Molecular profiling of lung cancer samples for activated oncogenes, including epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 (ROS1) and BRAF, is routinely performed to select the most appropriate up-front treatment. However, the identification of new therapeutic targets remains a high priority. Recently, MET exon 14 skipping mutations have emerged as novel actionable oncogenic alterations in NSCLC, sensitive to MET inhibition. In this review we discuss: (I) MET gene and MET receptor structure and signaling pathway; (II) MET exon 14 alterations; (III) current data on MET inhibitors, mainly focusing on selective MET tyrosine kinase inhibitors (TKIs), in the treatment of NSCLC with MET exon 14 skipping mutations. We identified the references for this review through a literature search of papers about MET, MET exon 14 skipping mutations, and MET inhibitors, published up to September 2020, by using PubMed, Scopus and Web of Science databases. We also searched on websites of main international cancer congresses (ASCO, ESMO, IASLC) for ongoing studies presented as abstracts. MET exon 14 skipping mutations have been associated with clinical activity of selective MET inhibitors, including capmatinib, that has recently received approval by FDA for clinical use in this subgroup of NSCLC patients. A large number of trials are testing MET inhibitors, also in combinatorial therapeutic strategies, in MET exon 14-altered NSCLC. Results from these trials are eagerly awaited to definitively establish the role and setting for use of these agents in NSCLC patients.
Collapse
Affiliation(s)
- Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Patology “G. Barresi”, University of Messina, Messina, Italy
| | - Marco Massafra
- Medical Oncology Unit, Department of Human Patology “G. Barresi”, University of Messina, Messina, Italy
| | - Vittorio Gebbia
- Medical Oncology and Supportive Care Unit, La Maddalena Cancer Center, Palermo, Italy;,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Antonio D’Aquino
- Medical Oncology Unit, Department of Human Patology “G. Barresi”, University of Messina, Messina, Italy
| | - Claudia Garipoli
- Medical Oncology Unit, Department of Human Patology “G. Barresi”, University of Messina, Messina, Italy
| | - Giuseppe Altavilla
- Medical Oncology Unit, Department of Human Patology “G. Barresi”, University of Messina, Messina, Italy
| | - Rafael Rosell
- Catalan Institute of Oncology, Laboratory of Cellular and Molecular Biology, Institute for Health Science Research Germans Trias i Pujol, Badalona, Barcelona, Spain;,Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital, Barcelona, Spain;,Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
50
|
Mori JI, Adachi K, Sakoda Y, Sasaki T, Goto S, Matsumoto H, Nagashima Y, Matsuyama H, Tamada K. Anti-tumor efficacy of human anti-c-met CAR-T cells against papillary renal cell carcinoma in an orthotopic model. Cancer Sci 2021; 112:1417-1428. [PMID: 33539630 PMCID: PMC8019206 DOI: 10.1111/cas.14835] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 01/01/2023] Open
Abstract
Chimeric antigen receptor (CAR)‐T cell therapy has shown salient efficacy in cancer immunotherapy, particularly in the treatment of B cell malignancies. However, the efficacy of CAR‐T for solid tumors remains inadequate. In this study, we displayed that c‐met is an appropriate therapeutic target for papillary renal cell carcinoma (PRCC) using clinical samples, developed an anti‐human c‐met CAR‐T cells, and investigated the anti‐tumor efficacy of the CAR‐T cells using an orthotopic mouse model as pre‐clinical research. Administration of the anti‐c‐met CAR‐T cells induced marked infiltration of the CAR‐T cells into the tumor tissue and unambiguous suppression of tumor growth. Furthermore, in combination with axitinib, the anti‐tumor efficacy of the CAR‐T cells was synergistically augmented. Taken together, our current study demonstrated the potential for clinical application of anti‐c‐met CAR‐T cells in the treatment of patients with PRCC.
Collapse
Affiliation(s)
- Jun-Ich Mori
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube, Japan.,Department of Urology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Keishi Adachi
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yukimi Sakoda
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Takahiro Sasaki
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube, Japan.,Department of Endocrinology, Metabolism, Hematological Science and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shunsuke Goto
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube, Japan.,Department of Urology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Hiroaki Matsumoto
- Department of Urology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yoji Nagashima
- Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hideyasu Matsuyama
- Department of Urology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Koji Tamada
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| |
Collapse
|