1
|
Yang J, Chen G, Wang R, Song C, Yi H. Navigating TAM receptor dynamics in tumour immunotherapy. Cancer Immunol Immunother 2025; 74:146. [PMID: 40088262 PMCID: PMC11910493 DOI: 10.1007/s00262-024-03879-z] [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: 07/02/2024] [Accepted: 11/02/2024] [Indexed: 03/17/2025]
Abstract
The TAM receptor family is getting more and more attention in the field of tumour immunity. Activation of TAM receptors not only aids in the survival and multiplication of tumour cells but also increases their likelihood of invading other cells and spreading. In addition, activation of TAM receptors helps to inhibit the anti-tumour immune response, allowing tumour cells to evade immune surveillance. In terms of therapeutic strategies, a number of inhibitors targeting TAM receptors are in preclinical and clinical development. Despite significant progress in clinical trials in recent years, challenges remain. This review delves into the kinetic characteristics of the TAM receptor family, their dual role in tumour immunity, and the transmission process of downstream signalling pathways. Based on this, we analysed and summarised the unique strategies and combination therapies for regulating tumour immunity using TAM receptor inhibitors. It not only helps to elucidate the key role of TAM receptors in tumour immunity but also provides new perspectives and strategies for future tumour therapy.
Collapse
Affiliation(s)
- Jihao Yang
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese, Medicine, Jinan, 250013, People's Republic of China
| | - Guanmin Chen
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese, Medicine, Jinan, 250013, People's Republic of China
| | - Rui Wang
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese, Medicine, Jinan, 250013, People's Republic of China
| | - Chengcheng Song
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese, Medicine, Jinan, 250013, People's Republic of China
| | - Huaqiang Yi
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese, Medicine, Jinan, 250013, People's Republic of China.
| |
Collapse
|
2
|
Zhang H, Wu LZ, Liu ZY, Jin ZB. Patient-derived induced pluripotent stem cells with a MERTK mutation exhibit cell junction abnormalities and aberrant cellular differentiation potential. World J Stem Cells 2024; 16:512-524. [PMID: 38817331 PMCID: PMC11135251 DOI: 10.4252/wjsc.v16.i5.512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/29/2024] [Accepted: 04/01/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND Human induced pluripotent stem cell (hiPSC) technology is a valuable tool for generating patient-specific stem cells, facilitating disease modeling, and investigating disease mechanisms. However, iPSCs carrying specific mutations may limit their clinical applications due to certain inherent characteristics. AIM To investigate the impact of MERTK mutations on hiPSCs and determine whether hiPSC-derived extracellular vesicles (EVs) influence anomalous cell junction and differentiation potential. METHODS We employed a non-integrating reprogramming technique to generate peripheral blood-derived hiPSCs with and hiPSCs without a MERTK mutation. Chromosomal karyotype analysis, flow cytometry, and immunofluorescent staining were utilized for hiPSC identification. Transcriptomics and proteomics were employed to elucidate the expression patterns associated with cell junction abnormalities and cellular differentiation potential. Additionally, EVs were isolated from the supernatant, and their RNA and protein cargos were examined to investigate the involvement of hiPSC-derived EVs in stem cell junction and differentiation. RESULTS The generated hiPSCs, both with and without a MERTK mutation, exhibited normal karyotype and expressed pluripotency markers; however, hiPSCs with a MERTK mutation demonstrated anomalous adhesion capability and differentiation potential, as confirmed by transcriptomic and proteomic profiling. Furthermore, hiPSC-derived EVs were involved in various biological processes, including cell junction and differentiation. CONCLUSION HiPSCs with a MERTK mutation displayed altered junction characteristics and aberrant differentiation potential. Furthermore, hiPSC-derived EVs played a regulatory role in various biological processes, including cell junction and differentiation.
Collapse
Affiliation(s)
- Hang Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Ling-Zi Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Zhen-Yu Liu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China.
| |
Collapse
|
3
|
Breton-Patient C, Billotte S, Duchambon P, Fontaine G, Bombard S, Piguel S. Light-Activatable Photocaged UNC2025 for Triggering TAM Kinase Inhibition in Bladder Cancer. Chembiochem 2024; 25:e202300855. [PMID: 38363151 DOI: 10.1002/cbic.202300855] [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: 12/20/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/17/2024]
Abstract
Photopharmacology is an emerging field that utilizes photo-responsive molecules to enable control over the activity of a drug using light. The aim is to limit the therapeutic action of a drug at the level of diseased tissues and organs. Considering the well-known implications of protein kinases in cancer and the therapeutic issues associated with protein kinase inhibitors, the photopharmacology is seen as an innovative and alternative solution with great potential in oncology. In this context, we developed the first photocaged TAM kinase inhibitors based on UNC2025, a first-in-class small molecule kinase inhibitor. These prodrugs showed good stability in biologically relevant buffer and rapid photorelease of the photoremovable protecting group upon UV-light irradiation (<10 min.). These light-activatable prodrugs led to a 16-fold decrease to a complete loss of kinase inhibition, depending on the protein and the position at which the coumarin-type phototrigger was introduced. The most promising candidate was the N,O-dicaged compound, showing the superiority of having two photolabile protecting groups on UNC2025 for being entirely inactive on TAM kinases. Under UV-light irradiation, the N,O-dicaged compound recovered its inhibitory potency in enzymatic assays and displayed excellent antiproliferative activity in RT112 cell lines.
Collapse
Affiliation(s)
- Chloé Breton-Patient
- Institut Curie, Université PSL CNRS UMR9187, Inserm U119, 91400, Orsay, France
- Université Paris-Saclay CNRS UMR9187, Inserm U119, 91400, Orsay, France
| | - Sébastien Billotte
- Université Paris-Saclay, Faculté de Pharmacie CNRS UMR 8076, 91400, Orsay, France
| | - Patricia Duchambon
- Institut Curie, Université PSL CNRS UMR9187, Inserm U119, 91400, Orsay, France
- Université Paris-Saclay CNRS UMR9187, Inserm U119, 91400, Orsay, France
| | - Gaëlle Fontaine
- Institut Curie, Université PSL CNRS UMR9187, Inserm U119, 91400, Orsay, France
- Université Paris-Saclay CNRS UMR9187, Inserm U119, 91400, Orsay, France
| | - Sophie Bombard
- Institut Curie, Université PSL CNRS UMR9187, Inserm U119, 91400, Orsay, France
- Université Paris-Saclay CNRS UMR9187, Inserm U119, 91400, Orsay, France
| | - Sandrine Piguel
- Université Paris-Saclay, Faculté de Pharmacie CNRS UMR 8076, 91400, Orsay, France
| |
Collapse
|
4
|
Ubil E, Zahid KR. Structure and functions of Mer, an innate immune checkpoint. Front Immunol 2023; 14:1244170. [PMID: 37936688 PMCID: PMC10626544 DOI: 10.3389/fimmu.2023.1244170] [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: 06/22/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Immunotherapy is a promising therapeutic tool that promotes the elimination of cancerous cells by a patient's own immune system. However, in the clinical setting, the number of cancer patients benefitting from immunotherapy is limited. Identification and targeting of other immune subsets, such as tumor-associated macrophages, and alternative immune checkpoints, like Mer, may further limit tumor progression and therapy resistance. In this review, we highlight the key roles of macrophage Mer signaling in immune suppression. We also summarize the role of pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes in tumor onset and progression and how Mer structure and activation can be targeted therapeutically to alter activation state. Preclinical and clinical studies focusing on Mer kinase inhibition have demonstrated the potential of targeting this innate immune checkpoint, leading to improved anti-tumor responses and patient outcomes.
Collapse
Affiliation(s)
- Eric Ubil
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | | |
Collapse
|
5
|
Gadiyar V, Patel G, Chen J, Vigil D, Ji N, Campbell V, Sharma K, Shi Y, Weiss MM, Birge RB, Davra V. Targeted degradation of MERTK and other TAM receptor paralogs by heterobifunctional targeted protein degraders. Front Immunol 2023; 14:1135373. [PMID: 37545504 PMCID: PMC10397400 DOI: 10.3389/fimmu.2023.1135373] [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: 12/31/2022] [Accepted: 05/26/2023] [Indexed: 08/08/2023] Open
Abstract
TAM receptors (TYRO3, AXL, and MERTK) comprise a family of homologous receptor tyrosine kinases (RTK) that are expressed across a range of liquid and solid tumors where they contribute to both oncogenic signaling to promote tumor proliferation and survival, as well as expressed on myeloid and immune cells where they function to suppress host anti-tumor immunity. In recent years, several strategies have been employed to inhibit TAM kinases, most notably small molecule tyrosine kinase inhibitors and inhibitory neutralizing monoclonal antibodies (mAbs) that block receptor dimerization. Targeted protein degraders (TPD) use the ubiquitin proteasome pathway to redirect E3 ubiquitin ligase activity and target specific proteins for degradation. Here we employ first-in-class TPDs specific for MERTK/TAMs that consist of a cereblon E3 ligase binder linked to a tyrosine kinase inhibitor targeting MERTK and/or AXL and TYRO3. A series of MERTK TPDs were designed and investigated for their capacity to selectively degrade MERTK chimeric receptors, reduce surface expression on primary efferocytic bone marrow-derived macrophages, and impact on functional reduction in efferocytosis (clearance of apoptotic cells). We demonstrate proof-of-concept and establish that TPDs can be tailored to either selectivity degrades MERTK or concurrently degrade multiple TAMs and modulate receptor expression in vitro and in vivo. This work demonstrates the utility of proteome editing, enabled by tool degraders developed here towards dissecting the therapeutically relevant pathway biology in preclinical models, and the ability for TPDs to degrade transmembrane proteins. These data also provide proof of concept that TPDs may serve as a viable therapeutic strategy for targeting MERTK and other TAMs and that this technology could be expanded to other therapeutically relevant transmembrane proteins.
Collapse
Affiliation(s)
- Varsha Gadiyar
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, NJ, United States
| | - Gopi Patel
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, NJ, United States
| | - Jesse Chen
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Dominico Vigil
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Nan Ji
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Veronica Campbell
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Kirti Sharma
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Yatao Shi
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Matthew M. Weiss
- Department of Research and Development, Kymera Therapeutics, Watertown, MA, United States
| | - Raymond B. Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, NJ, United States
| | - Viralkumar Davra
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
6
|
Kharouf N, Flanagan TW, Hassan SY, Shalaby H, Khabaz M, Hassan SL, Megahed M, Haikel Y, Santourlidis S, Hassan M. Tumor Microenvironment as a Therapeutic Target in Melanoma Treatment. Cancers (Basel) 2023; 15:3147. [PMID: 37370757 DOI: 10.3390/cancers15123147] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The role of the tumor microenvironment in tumor growth and therapy has recently attracted more attention in research and drug development. The ability of the microenvironment to trigger tumor maintenance, progression, and resistance is the main cause for treatment failure and tumor relapse. Accumulated evidence indicates that the maintenance and progression of tumor cells is determined by components of the microenvironment, which include stromal cells (endothelial cells, fibroblasts, mesenchymal stem cells, and immune cells), extracellular matrix (ECM), and soluble molecules (chemokines, cytokines, growth factors, and extracellular vesicles). As a solid tumor, melanoma is not only a tumor mass of monolithic tumor cells, but it also contains supporting stroma, ECM, and soluble molecules. Melanoma cells are continuously in interaction with the components of the microenvironment. In the present review, we focus on the role of the tumor microenvironment components in the modulation of tumor progression and treatment resistance as well as the impact of the tumor microenvironment as a therapeutic target in melanoma.
Collapse
Affiliation(s)
- Naji Kharouf
- Biomaterials and Bioengineering, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Unité Mixte de Recherche 1121, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Thomas W Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | - Sofie-Yasmin Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Dusseldorf, Germany
| | - Hosam Shalaby
- Department of Urology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Marla Khabaz
- Department of Production, Beta Factory for Veterinary Pharmaceutical Industries, Damascus 0100, Syria
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Dusseldorf, Germany
| | - Mosaad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany
| | - Youssef Haikel
- Biomaterials and Bioengineering, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Unité Mixte de Recherche 1121, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Simeon Santourlidis
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Mohamed Hassan
- Biomaterials and Bioengineering, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Unité Mixte de Recherche 1121, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
7
|
Kostecki KL, Iida M, Wiley AL, Kimani S, Mehall B, Tetreault K, Alexandridis R, Yu M, Hong S, Salgia R, Bruce JY, Birge RB, Harari P, Wheeler DL. Dual Axl/MerTK inhibitor INCB081776 creates a proinflammatory tumor immune microenvironment and enhances anti-PDL1 efficacy in head and neck cancer. Head Neck 2023; 45:1255-1271. [PMID: 36939040 PMCID: PMC10079616 DOI: 10.1002/hed.27340] [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: 10/31/2022] [Revised: 02/01/2023] [Accepted: 02/22/2023] [Indexed: 03/21/2023] Open
Abstract
BACKGROUND The tyrosine kinase receptors Axl and MerTK are highly overexpressed in head and neck cancer (HNC) cells, where they are critical drivers of survival, proliferation, metastasis, and therapeutic resistance. METHODS We investigated the role of Axl and MerTK in creating an immunologically "cold" tumor immune microenvironment (TIME) by targeting both receptors simultaneously with a small molecule inhibitor of Axl and MerTK (INCB081776). Effects of INCB081776 and/or anti-PDL1 on mouse oral cancer (MOC) cell growth and on the TIME were evaluated. RESULTS Targeting Axl and MerTK can reduce M2 and induce M1 macrophage polarization. In vivo, INCB081776 treatment alone or with anti-PDL1 appears to slow MOC tumor growth, increase proinflammatory immune infiltration, and decrease anti-inflammatory immune infiltration. CONCLUSIONS This data indicates that simultaneous targeting of Axl and MerTK with INCB081776, either alone or in combination with anti-PDL1, slows tumor growth and creates a proinflammatory TIME in mouse models of HNC.
Collapse
Affiliation(s)
- Kourtney L Kostecki
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Mari Iida
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Anne L Wiley
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Stanley Kimani
- Rutgers Biomedical Health and Sciences, Rutgers University, Newark, NJ, USA
| | - Bridget Mehall
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kaitlin Tetreault
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Roxana Alexandridis
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Menggang Yu
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, University of Wisconsin School of Pharmacy, Madison, WI, USA
- Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul, Korea
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Ravi Salgia
- Department of Medical Oncology and Experimental Therapeutics, Comprehensive Cancer Center, City of Hope, Duarte, CA, USA
| | - Justine Y Bruce
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Raymond B Birge
- Rutgers Biomedical Health and Sciences, Rutgers University, Newark, NJ, USA
| | - Paul Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Deric L Wheeler
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
8
|
The First-In-Class Anti-AXL×CD3ε Pronectin™-Based Bispecific T-Cell Engager Is Active in Preclinical Models of Human Soft Tissue and Bone Sarcomas. Cancers (Basel) 2023; 15:cancers15061647. [PMID: 36980534 PMCID: PMC10046451 DOI: 10.3390/cancers15061647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
Sarcomas are heterogeneous malignancies with limited therapeutic options and a poor prognosis. We developed an innovative immunotherapeutic agent, a first-in-class Pronectin™-based Bispecific T-Cell Engager (pAXL×CD3ε), for the targeting of AXL, a TAM family tyrosine kinase receptor highly expressed in sarcomas. AXL expression was first analyzed by flow cytometry, qRT-PCR, and Western blot on a panel of sarcoma cell lines. The T-cell-mediated pAXL×CD3ε cytotoxicity against sarcoma cells was investigated by flow cytometry, luminescence assay, and fluorescent microscopy imaging. The activation and degranulation of T cells induced by pAXL×CD3ε were evaluated by flow cytometry. The antitumor activity induced by pAXL×CD3ε in combination with trabectedin was also investigated. In vivo activity studies of pAXL×CD3ε were performed in immunocompromised mice (NSG), engrafted with human sarcoma cells and reconstituted with human peripheral blood mononuclear cells from healthy donors. Most sarcoma cells showed high expression of AXL. pAXL×CD3ε triggered T-lymphocyte activation and induced dose-dependent T-cell-mediated cytotoxicity. The combination of pAXL×CD3ε with trabectedin increased cytotoxicity. pAXL×CD3ε inhibited the in vivo growth of human sarcoma xenografts, increasing the survival of treated mice. Our data demonstrate the antitumor efficacy of pAXL×CD3ε against sarcoma cells, providing a translational framework for the clinical development of pAXL×CD3ε in the treatment of human sarcomas, aggressive and still-incurable malignancies.
Collapse
|
9
|
Wu S, Liao M, Li M, Sun M, Xi N, Zeng Y. Structure-based discovery of potent inhibitors of Axl: design, synthesis, and biological evaluation. RSC Med Chem 2022; 13:1246-1264. [PMID: 36325401 PMCID: PMC9579923 DOI: 10.1039/d2md00153e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/12/2022] [Indexed: 07/22/2023] Open
Abstract
Commonly overexpressed in many cancers and associated with tumor growth, metastasis, drug resistance, and poor overall survival, Axl has emerged as a promising target for cancer therapy. However, the availability of new chemical forms for Axl inhibition is limited. Herein, we present the development and characterization of novel Axl inhibitors, including the design, synthesis, and structure-activity relationships (SARs) of a series of diphenylpyrimidine-diamine derivatives. Most of these compounds exhibited remarkable activity against the Axl kinase. In particular, the promising compound m16 showed the highest enzymatic inhibitory potency (IC50 = 5 nM) and blocked multiple tumor cells' proliferation potencies (the CC50 of 4 out of 42 cancer cell lines <100 nM). Furthermore, compound m16 also possessed preferable pharmacokinetic profiles and liver microsome stability. All these favorable results make m16 a good leading therapeutic candidate for further development.
Collapse
Affiliation(s)
- Shuang Wu
- Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), Hunan Normal University Changsha 410081 PR China
- Ningbo University School of Medicine 818 Fenghua Road Ningbo Zhejiang 315211 China
- Sunshine Lake Pharmaceutical Co. Ltd Dongyangguang Hi-tech Park Dongguan Guangdong 523871 China
| | - Min Liao
- Sunshine Lake Pharmaceutical Co. Ltd Dongyangguang Hi-tech Park Dongguan Guangdong 523871 China
- School of Chemistry & Chemical Engineering, Guangxi University Nanning 530004 China
| | - Minxiong Li
- Sunshine Lake Pharmaceutical Co. Ltd Dongyangguang Hi-tech Park Dongguan Guangdong 523871 China
| | - Mingming Sun
- Ningbo University School of Medicine 818 Fenghua Road Ningbo Zhejiang 315211 China
- Sunshine Lake Pharmaceutical Co. Ltd Dongyangguang Hi-tech Park Dongguan Guangdong 523871 China
| | - Ning Xi
- Ningbo University School of Medicine 818 Fenghua Road Ningbo Zhejiang 315211 China
- Sunshine Lake Pharmaceutical Co. Ltd Dongyangguang Hi-tech Park Dongguan Guangdong 523871 China
| | - Youlin Zeng
- Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), Hunan Normal University Changsha 410081 PR China
| |
Collapse
|