1
|
Hmedat ANA, Doondeea J, Ebner D, Feller SM, Lewitzky M. The Src family kinase inhibitor drug Dasatinib and glucocorticoids display synergistic activity against tongue squamous cell carcinoma and reduce MET kinase activity. Cell Commun Signal 2025; 23:293. [PMID: 40537792 DOI: 10.1186/s12964-025-02129-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/25/2025] [Indexed: 06/22/2025] Open
Abstract
BACKGROUND Tongue squamous cell carcinoma (TSCC) is an aggressive cancer associated with a poor prognosis and limited treatment options, necessitating new drug targets to improve therapeutic outcomes. Our current work studies protein tyrosine kinases as well-known targets for successful cancer therapies. It focuses on Src family kinases (SFK), which are known to play a critical role in some head and neck tumors. METHODS Western blot analyses of phospho-tyrosine protein patterns in 34 TSCC lines facilitated the investigation of SFK as contributors to these phosphorylations. The SFK inhibitors PP2 and Dasatinib were utilized to determine SFK contributions to cell motility and survival. A high-throughput screen with 1600 FDA-approved drugs was performed with three TSCC lines to discover drugs that act synergistically with Dasatinib against TSCC cell viability. Glucocorticoids emerged as potential candidates and were further investigated in 2D culture and by 3D soft agar colony formation. Dexamethasone was chosen as the major tool for our analyses of synergistic effects of Dasatinib and glucocorticoids on TSCC lines. Effects on the cell cycle were investigated by flow cytometry and expression levels of cell cycle regulators. Senescence was analyzed by senescence-associated β galactosidase detection and p27Kip1 protein expression. Autophagy was measured by Acridine Orange staining. RESULTS A panel of 34 TSCC lines showed a surprisingly homogenous pTyr-protein pattern and a prominent 130 kDa pTyr-protein. Inhibition of SFK activity greatly reduced overall pTyr-protein levels and p130Cas tyrosine phosphorylation. It also impaired TSCC viability in 2D cell culture and 3D soft agar colony formation. A high-throughput drug combination screen with Dasatinib identified glucocorticoids as promising candidates for synergistic activity. Dasatinib and Dexamethasone combination treatment showed strong synergistic effects on Src and p130Cas phosphorylation and led to reduced p130Cas expression. Dexamethasone also suppressed phosphorylation of the MET kinase and its key substrate Gab1. On the cellular level, Dasatinib combination with glucocorticoids led to G1 cell cycle arrest, appeared to increase senescence and enhanced autophagy. This was also reflected by effects on cell cycle regulatory proteins, including CDKs and cyclins. CONCLUSION This work is the first to show a strong synergistic activity of Dasatinib in combination with clinically used glucocorticoids in solid tumors. Furthermore, the tyrosine kinase MET and its effector protein Gab1 are newly identified glucocorticoid targets. Given the extensive research on MET as a drug target in various cancers, our findings have the potential to advance future cancer treatments.
Collapse
Affiliation(s)
- Ali N A Hmedat
- Institute of Molecular Medicine, Medical Faculty of the Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Jessica Doondeea
- Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Daniel Ebner
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Stephan M Feller
- Institute of Molecular Medicine, Medical Faculty of the Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Marc Lewitzky
- Institute of Molecular Medicine, Medical Faculty of the Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany.
| |
Collapse
|
2
|
Russo G, Scimone C, Palumbo L, Roscigno G, Sarracino C, Tomaiuolo I, Pisapia P, Pepe F, Rocco D, Gridelli C, Troncone G, Malapelle U. Biologics for novel driver altered non-small cell lung cancer: potential and pitfalls. Crit Rev Oncol Hematol 2025; 212:104748. [PMID: 40324663 DOI: 10.1016/j.critrevonc.2025.104748] [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: 03/06/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
Precision medicine has revolutionized clinical paradigm of lung cancer (LC) patients optimizing therapeutical options on the basis of molecular fingerprinting of tumor cells. The advent of the genomic era contributed to the widespread diffusion of sequencing technologies laying the basis for the approval of an increasing number of clinically relevant predictive biomarkers in clinical settings. In the rapidly evolving scenario of predictive biomarkers, mandatory testing genes demonstrated a statistically significant clinical benefit in LC patients elected to molecular tests, but emerging biomarkers are under investigation to raise the bar in the clinical management of LC patients. To date, promising IHC-based predictive biomarkers emerged as potentially integrative tools in the panel of clinically approved biomarkers. On this basis, genomic, transcriptomic and proteomic data are gaining ground toward "3D" biology" supporting the need of a multidimensional analysis of tumor cells to clinically stratify LC patients. Here we sought to overview the most promising biomarkers investigated in clinical trials to be integrated into diagnostic panel of predictive biomarkers tools for NSCLC patients.
Collapse
Affiliation(s)
- Gianluca Russo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Claudia Scimone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Lucia Palumbo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Claudia Sarracino
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Ilaria Tomaiuolo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Danilo Rocco
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, Napoli, Italy
| | - Cesare Gridelli
- Division of Medical Oncology, 'S. G. Moscati' Hospital, Avellino, Italy
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy.
| |
Collapse
|
3
|
Wang M, Ma Q, Suthe SR, Hudson RE, Pan JY, Mikelis C, Zhu MJ, Wu ZG, Shi DR, Yao HP. Humanized dual-targeting antibody-drug conjugates specific to MET and RON receptors as a pharmaceutical strategy for the treatment of cancers exhibiting phenotypic heterogeneity. Acta Pharmacol Sin 2025; 46:1375-1389. [PMID: 39837982 PMCID: PMC12032285 DOI: 10.1038/s41401-024-01458-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025]
Abstract
Cancer heterogeneity, characterized by diverse populations of tumorigenic cells, involves the occurrence of differential phenotypes with variable expressions of receptor tyrosine kinases. Aberrant expressions of mesenchymal-epithelial transition (MET) and recepteur d'origine nantais (RON) receptors contribute to the phenotypic heterogeneity of cancer cells, which poses a major therapeutic challenge. This study aims to develop a dual-targeting antibody-drug conjugate (ADC) that can act against both MET and RON for treating cancers with high phenotypic heterogeneity. Through immunohistochemical staining, we show that MET and RON expressions are highly heterogeneous with differential combinations in more than 40% of pancreatic and triple-negative breast cancer cases. This expressional heterogeneity provides the rationale to target both receptors for cancer therapy. A humanized bispecific monoclonal antibody specific to both MET and RON (PCMbs-MR) is generated through IgG recombination using monoclonal antibody sequences specific to MET and RON, respectively. Monomethyl auristatin E is conjugated to PCMbs-MR to generate a dual-targeting ADC (PCMdt-MMAE), with a drug-to-antibody ratio of 4:1. Various cancer cell lines were used to determine PCMdt-MMAE-mediated biological activities. The efficacy of PCMdt-MMAE in vivo is evaluated using multiple xenograft tumor models. PCMdt-MMAE shows a favorable pharmacokinetic profile, with a maximum tolerated dose of ~30 mg/kg in mice. Toxicological studies using Sprague-Dawley rats reveal that PCMdt-MMAE is relatively safe with slight-to-moderate, temporary, and reversible adverse events. Functionally, PCMdt-MMAE induces a robust internalization of both MET and RON and causes a large-scale cell death in cancer cell lines exhibiting MET and RON heterogeneous co-expressions. Both in vitro and in vivo studies demonstrate that the dual-targeting approach in the form of an ADC is highly effective with a long-lasting effect against tumors exhibiting MET/RON heterogeneous phenotypes. Hence, we can suggest that a dual-targeting ADC specific to both MET and RON can be employed as a novel therapeutic strategy for tumors with expressional phenotypic heterogeneity.
Collapse
Affiliation(s)
- Minghai Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, 79106, TX, USA
- Cancer Biology Research Center, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, 79106, TX, USA
| | - Qi Ma
- Comprehensive Genitourinary Cancer Center, First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
- Translational Research Laboratory for Urology, The Key Laboratory of Ningbo City, Ningbo, 315000, China
| | - Sreedhar Reddy Suthe
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, 79106, TX, USA
| | - Rachel E Hudson
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, 79106, TX, USA
| | - Jing-Ying Pan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Constantinos Mikelis
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, 79106, TX, USA
- Cancer Biology Research Center, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, 79106, TX, USA
| | - Miao-Jin Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhi-Gang Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Dan-Rong Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Hang-Ping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
4
|
Liu X, Chen Y, Zhang F, Qiu F, Xu X, Zhang J, He S, Ding D, Tan W, Song S. Preclinical Evaluation of 68Ga-Labeled SL1 Aptamer for c-Met Targeted PET Imaging. Mol Pharm 2025; 22:1615-1623. [PMID: 39930702 DOI: 10.1021/acs.molpharmaceut.4c01344] [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] [Indexed: 03/04/2025]
Abstract
Tyrosine protein kinase c-Met, encoded by the Met gene, is a membrane-associated receptor tyrosine kinase that is often aberrantly expressed in a wide range of tumors. The development of imaging probes specifically targeting c-Met is critical for improving cancer diagnostics. In this study, we successfully designed and fabricated an aptamer molecular imaging probe ([68Ga]Ga-NOTA-SL1) with high radiochemical purity (RCP), good stability in vitro, and high affinity for c-Met expressed tumors. As shown by the micro-PET/CT scanning, [68Ga]Ga-NOTA-SL1 efficiently imaged tumor models with varying c-Met expression. The quantitative analysis of micro-PET/CT showed tumor uptake of [68Ga]Ga-NOTA-SL1 in the HCC827 tumor models (30 min, 2.93 ± 0.64%ID/g; 60 min, 2.03 ± 0.67%ID/g; 90 min, 1.63 ± 0.61%ID/g), PC-9 tumor models (30 min, 2.1 ± 0.72%ID/g; 60 min, 1.7 ± 0.56%ID/g; 90 min, 1.33 ± 0.38%ID/g), and HCT116 tumor models (30 min, 1.4 ± 0.17%ID/g; 60 min, 1.23 ± 0.15%ID/g; 90 min, 0.97 ± 0.21%ID/g). The results of immunohistochemistry (IHC) further confirmed the targeting ability of [68Ga]Ga-NOTA-SL1 to c-Met from a molecular pathological perspective. The probe effectively imaged c-Met-positive tumors and demonstrated a favorable metabolism profile and targeting performance in non-small cell lung cancer (NSCLC) or colorectal cancer tumor models. Consequently, this probe shows promise as an imaging agent capable of providing valuable diagnostic insights into tumors with aberrant c-Met expression.
Collapse
Affiliation(s)
- Xuwei Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai 200233, P. R. China
| | - Yamei Chen
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Fengsheng Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| | - Fengshuang Qiu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| | - Xiaoping Xu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| | - Simin He
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| | - Ding Ding
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College and Center for Biomedical Imaging, Fudan University, Shanghai 200032, P. R. China
- Center for Biomedical Imaging, Fudan University, Shanghai 200032, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai 200032, P. R. China
- Key Laboratory of Nuclear Physics and Ion-Beam Application (MOE), Fudan University, Shanghai 200433, China
- Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| |
Collapse
|
5
|
Tripathy A, Corkos P, Blouw B, Montgomery DA, Moore M, Hedrick MH, Youssef M, Kumthekar PU. Longitudinal CSF Tumor Cell Enumeration and Mutational Analysis as a Driver for Leptomeningeal Disease Management. Cancers (Basel) 2025; 17:825. [PMID: 40075672 PMCID: PMC11899081 DOI: 10.3390/cancers17050825] [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: 01/03/2025] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Leptomeningeal disease (LMD) is challenging to diagnose and monitor given the poor sensitivity of current gold-standard diagnostics. Cerebrospinal fluid tumor cells (CSF-TCs) have been studied as a biomarker for disease management because oncogene amplification of the primary, metastatic, and CNS metastatic tumors can be heterogeneous. The CNSide platform enumerates CSF-TCs and analyzes oncogene expression via immunocytochemistry (ICC), fluorescent in situ hybridization (FISH), and next-generation sequencing (NGS). We report the utility of this combined enumerative and mutational testing for LMD diagnosis and disease monitoring. METHODS A multicenter, retrospective analysis of commercially ordered assays from two health systems between January 2020 and July 2023 included 613 tests on 218 individual patients with suspected or confirmed LMD. To date, this is the largest cohort of patients in LMD literature evaluated using CSF-TCs. RESULTS CSF-TCs were detected in 67% (412/613) of samples. The most analyzed cancer types were breast (n = 105) and lung (n = 65). In lung cancer, anaplastic lymphoma kinase (ALK) was detected in 14% (17/118), and c-MET was detected in 61% (78/128). In breast cancer, HER2 was detected in 39% (65/168), and estrogen receptor (ER) was detected in 26% (44/168). Sixty-six patients underwent 2+ longitudinal CSF draws; among these, there were 58 flips in oncogene detection over time, and 30% (20/66) of patients had at least one biomarker change in the CSF. CONCLUSIONS Longitudinal combined ICC/FISH/NGS CSF testing demonstrates a wide range in CSF-TC enumeration, which may be correlated with clinical course, and furthermore identifies actionable tumor markers that frequently fluctuate over time. Utilization of this platform would enable timely, personalized LMD-specific chemotherapy.
Collapse
Affiliation(s)
- Arushi Tripathy
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | - Michael Youssef
- Department of Neurology, University of Southwestern Medical Center, Dallas, TX 75390, USA
| | - Priya U. Kumthekar
- Neurology (Neuro-Oncology) and Medicine (Hematology and Oncology), Northwestern Medicine Lou and Jean Malani Brain Tumor Institute, Chicago, IL 60611, USA
| |
Collapse
|
6
|
He H, Deng X, Wang Z, Chen J. Recent progress in the development of peptide-drug conjugates (PDCs) for cancer therapy. Eur J Med Chem 2025; 284:117204. [PMID: 39731788 DOI: 10.1016/j.ejmech.2024.117204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/10/2024] [Accepted: 12/22/2024] [Indexed: 12/30/2024]
Abstract
Peptide-drug conjugates (PDCs) are emerging therapeutic agents composed of peptides, linkers, and payloads, which possess favorable targeting capability and can deliver enough payloads to the tumor sites with minimized impact on healthy tissues. However, only a few PDCs have been approved for clinical use so far. To advance the research on PDCs, this review summarizes the approved PDCs, and PDCs in clinical and preclinical stages based on the payload types. Additionally, the biological activity and pharmacokinetic properties of preclinical PDCs are detailedly described. Lastly, the challenges and future development directions of PDCs are discussed. This review aims to inspire insights into the development of PDCs for cancer treatment.
Collapse
Affiliation(s)
- Haiqi He
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin Deng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinic Al Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhijie Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Shenzhen Key Laboratory of Viral Oncology, Ministry of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
7
|
Singh S, Nigam V, Kasana S, Kurmi BD, Gupta GD, Patel P. Targeting c-Met in Cancer Therapy: Unravelling Structure-activity Relationships and Docking Insights for Enhanced Anticancer Drug Design. Curr Top Med Chem 2025; 25:409-433. [PMID: 39484763 DOI: 10.2174/0115680266331025241015084546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/02/2024] [Accepted: 09/28/2024] [Indexed: 11/03/2024]
Abstract
The c-Met receptor, a pivotal player in oncogenesis and tumor progression, has become a compelling target for anticancer drug development. This review explores the intricate landscape of Structure-Activity Relationship (SAR) studies and molecular binding analyses performed on c-Met inhibitors. Through a comprehensive examination of various chemical scaffolds and modifications, SAR investigations have elucidated critical molecular features essential for the potent inhibition of c-Met activity. Additionally, molecular docking studies have provided invaluable insights into how c-Met inhibitors interact with their target receptor, facilitating the rational design of novel compounds with enhanced efficacy and selectivity. This review highlights key findings from recent SAR and docking studies, particularly focusing on the structural determinants that govern inhibition potency and selectivity. Furthermore, the integration of computational methodologies with experimental approaches has accelerated the discovery and optimization of c-Met inhibitors, fostering the advancement of promising candidates for clinical applications. Overall, this review underscores the pivotal role of SAR and molecular docking studies in advancing our understanding of c-Met inhibition and guiding the rational design of next-generation anticancer agents targeting this pathway.
Collapse
Affiliation(s)
- Surbhi Singh
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Vaibhav Nigam
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Shivani Kasana
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| |
Collapse
|
8
|
Salarinejad S, Seyfi S, Hayashi S, Moghimi S, Toolabi M, Taslimi P, Firoozpour L, Usui T, Foroumadi A. Design, synthesis, and biological evaluation of new biaryl derivatives of cycloalkyl diacetamide bearing chalcone moiety as type II c-MET kinase inhibitors. Mol Divers 2024; 28:4167-4180. [PMID: 38466553 DOI: 10.1007/s11030-024-10807-x] [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: 06/27/2023] [Accepted: 01/05/2024] [Indexed: 03/13/2024]
Abstract
Many human cancers have been associated with the deregulation of the mesenchymal-epithelial transition factor tyrosine kinase (MET) receptor, a promising drug target for anticancer drug discovery. Herein, we report the discovery of a novel structure of potent chalcone-based derivatives type II c-Met inhibitors which are comparable to Foretinib (IC50 = 14 nM) as a potent reference drug. Based on our design strategy, we also expected an anti-tubulin activity for the compounds. However, the weak inhibitory effects on microtubules were confirmed by cell cycle analyses implicated that the observed cytotoxicity against HeLa cells probably was not derived from tubulin inhibition. Compounds 14q and 14k with IC50 values of 24 nM and 45 nM, respectively, demonstrated favorable inhibition of MET kinase activity, and desirable bonding interactions in the ligand-MET enzyme complex stability in molecular docking studies.
Collapse
Affiliation(s)
- Somayeh Salarinejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheila Seyfi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Seiko Hayashi
- Institute of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan
| | - Setareh Moghimi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Toolabi
- Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Parham Taslimi
- Department of Biotechnology, Faculty of Science, Bartin University, 74100, Bartin, Turkey
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Takeo Usui
- Institute of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan.
| | - Alireza Foroumadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Park JH, Shin SJ, Kim HJ, Oh S, Cho YM. Histopathologic classification and immunohistochemical features of papillary renal neoplasm with potential therapeutic targets. J Pathol Transl Med 2024; 58:321-330. [PMID: 39257049 PMCID: PMC11573472 DOI: 10.4132/jptm.2024.07.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Papillary renal cell carcinoma (pRCC) is the second most common histological subtype of renal cell carcinoma and is considered a morphologically and molecularly heterogeneous tumor. Accurate classification and assessment of the immunohistochemical features of possible therapeutic targets are needed for precise patient care. We aimed to evaluate immunohistochemical features and possible therapeutic targets of papillary renal neoplasms. METHODS We collected 140 papillary renal neoplasms from three different hospitals and conducted immunohistochemical studies on tissue microarray slides. We performed succinate dehydrogenase B, fumarate hydratase, and transcription factor E3 immunohistochemical studies for differential diagnosis and re-classified five cases (3.6%) of papillary renal neoplasms. In addition, we conducted c-MET, p16, c-Myc, Ki-67, p53, and stimulator of interferon genes (STING) immunohistochemical studies to evaluate their pathogenesis and value for therapeutic targets. RESULTS We found that c-MET expression was more common in pRCC (classic) (p = .021) among papillary renal neoplasms and Ki-67 proliferation index was higher in pRCC (not otherwise specified, NOS) compared to that of pRCC (classic) and papillary neoplasm with reverse polarity (marginal significance, p = .080). Small subsets of cases with p16 block positivity (4.5%) (pRCC [NOS] only) and c-Myc expression (7.1%) (pRCC [classic] only) were found. Also, there were some cases showing STING expression and those cases were associated with increased Ki-67 proliferation index (marginal significance, p = .063). CONCLUSIONS Our findings suggested that there are subsets of pRCC with c-MET, p16, c-MYC, and STING expression and those cases could be potential candidates for targeted therapy.
Collapse
Affiliation(s)
- Jeong Hwan Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Su-Jin Shin
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun-Jung Kim
- Department of Pathology, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Sohee Oh
- Medical Research Collaborating Center, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
10
|
Chilamakuri R, Agarwal S. Repurposing of c-MET Inhibitor Tivantinib Inhibits Pediatric Neuroblastoma Cellular Growth. Pharmaceuticals (Basel) 2024; 17:1350. [PMID: 39458991 PMCID: PMC11510580 DOI: 10.3390/ph17101350] [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: 08/28/2024] [Revised: 09/22/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Dysregulation of receptor tyrosine kinase c-MET is known to promote tumor development by stimulating oncogenic signaling pathways in different cancers, including pediatric neuroblastoma (NB). NB is an extracranial solid pediatric cancer that accounts for almost 15% of all pediatric cancer-related deaths, with less than a 50% long-term survival rate. Results: In this study, we analyzed a large cohort of primary NB patient data and revealed that high MET expression strongly correlates with poor overall survival, disease progression, relapse, and high MYCN levels in NB patients. To determine the effects of c-MET in NB, we repurposed a small molecule inhibitor, tivantinib, and found that c-MET inhibition significantly inhibits NB cellular growth. Tivantinib significantly blocks NB cell proliferation and 3D spheroid tumor formation and growth in different MYCN-amplified and MYCN-non-amplified NB cell lines. Furthermore, tivantinib blocks the cell cycle at the G2/M phase transition and induces apoptosis in different NB cell lines. As expected, c-MET inhibition by tivantinib inhibits the expression of multiple genes in PI3K, STAT, and Ras cell signaling pathways. Conclusions: Overall, our data indicate that c-MET directly regulates NB growth and 3D spheroid growth, and c-MET inhibition by tivantinib may be an effective therapeutic approach for high-risk NB. Further developing c-MET targeted therapeutic approaches and combining them with current therapies may pave the way for effectively translating novel therapies for NB and other c-MET-driven cancers.
Collapse
Affiliation(s)
| | - Saurabh Agarwal
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY 11439, USA
| |
Collapse
|
11
|
Mobaraki S, Nissen PH, Donskov F, Wozniak A, Van Herck Y, Coosemans L, van Nieuwenhuyse T, Lambrechts D, Bechter O, Baldewijns M, Roussel E, Laenen A, Beuselinck B. Cabozantinib Induces Isolated Hyperbilirubinemia in Renal Cell Carcinoma Patients carrying the UGT1A1*28 Polymorphism. Clin Genitourin Cancer 2024; 22:102180. [PMID: 39155162 DOI: 10.1016/j.clgc.2024.102180] [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: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Genetic variants of UGT1A1, involved in glucuronidation and clearance of bilirubin, are associated with reduced bilirubin metabolization and drug-induced isolated hyperbilirubinemia. We studied the impact of the UGT1A1*28 polymorphism on drug-induced isolated hyperbilirubinemia in metastatic renal cell carcinoma patients treated with pazopanib, cabozantinib, and axitinib. METHODS We genotyped the UGT1A1*28 TA6/TA6-TA6/TA7-TA7/TA7 polymorphism and correlated with median baseline, on-treatment and peak bilirubin levels during therapy, incidence of grade-1- or -2 (G1/2)-hyperbilirubinemia and time-to-G1-hyperbilirubinemia. RESULTS Of the 66 patients treated with pazopanib, 29 received axitinib and 28 cabozantinib upon progression. Median baseline bilirubin was higher in TA7/TA7-carriers versus TA6/TA6+TA6/TA7-carriers at start of pazopanib (P < .0001), cabozantinib (P < .0001), and axitinib (P = .007). During pazopanib therapy, median bilirubin increased 1.4-fold in TA7/TA7+TA6/TA7-carriers but not in TA6/TA6-carriers. On cabozantinib, bilirubin increased 1.5-fold in TA7/TA7-carriers but not in TA6/TA6+TA6/TA7-carriers. Axitinib did not increase bilirubin in any genotype. Peak bilirubin in TA7/TA7- versus TA6/TA6+TA6/TA7-carriers was higher on pazopanib (P < .0001) or cabozantinib (P < .0001). With pazopanib, G1-hyperbilirubinemia occurred in 57% of TA7/TA7- and 12% of TA6/TA6+TA6/TA7-carriers (P = .0009) and G2-hyperbilirubinemia in 36% and 6% of the patients, respectively (P = .004). On cabozantinib, G1-hyperbilirubinemia occurred in 100% of TA7/TA7- and 5% of TA6/TA6+TA6/TA7-carriers (P < .0001) and G2-hyperbilirubinemia in 33% and 0% of the patients, respectively (P = .04). On axitinib, no correlation between the genotypes and G1/2-hyperbilirubinemia was observed. CONCLUSION We validate the previously described impact of the UGT1A1*28 polymorphism on isolated bilirubin increase on pazopanib. We report for the first time that cabozantinib also interferes with UGT1A1 and causes isolated bilirubin increase.
Collapse
Affiliation(s)
- Sajedeh Mobaraki
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Peter Henrik Nissen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Frede Donskov
- Department of Oncology, University Hospital of Southern Denmark, Esbjerg, Denmark
| | | | - Yannick Van Herck
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Lina Coosemans
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | | | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Oliver Bechter
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | | | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | | | - Benoit Beuselinck
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
12
|
Chen Y, Morihiro K, Nemoto Y, Ichimura A, Ueki R, Sando S, Okamoto A. Selective inhibition of cancer cell migration using a pH-responsive nucleobase-modified DNA aptamer. Chem Sci 2024:d4sc04424j. [PMID: 39355222 PMCID: PMC11440363 DOI: 10.1039/d4sc04424j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
Because of the extracellular acidic microenvironment of cancer cells, many pH-responsive molecules have become indispensable materials for bioanalysis and targeted therapy development. pH-Responsive DNA aptamers, which selectively bind to target proteins in cancer cells, have become a key research target in the therapeutic field. However, conventional pH-responsive aptamers have fatal drawbacks, such as complex structures, sequence limitation, and difficulties in mass production, as they require special nucleic acid structures, including the i-motif and DNA triplex. To address these issues, we utilized AnC, which is an unnatural nucleobase with a pK aH of 5.9, to construct a simple pH-responsive DNA aptamer (CSL1-II) for selective binding to the c-Met protein expressed in cancer cells. CSL1-II in a weakly acidic environment had a stronger inhibitory effect on the HGF/c-Met pathway and exerted a strong controlling effect on the spreading and migration of cancer cells. Our strategy provides a simple and versatile method to develop pH-responsive DNA aptamers and represents the first example of a cancer-selective c-Met antagonist that inhibits cell migration.
Collapse
Affiliation(s)
- Yuyuan Chen
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-8656 Japan
| | - Kunihiko Morihiro
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-8656 Japan
| | - Yui Nemoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-8656 Japan
| | - Akito Ichimura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-8656 Japan
| | - Ryosuke Ueki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-8656 Japan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-8656 Japan
| | - Akimitsu Okamoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo 7-3-1 Hongo Bunkyo-ku Tokyo 113-8656 Japan
| |
Collapse
|
13
|
Bumpers QA, Pipal RW, Benz-Weeden AM, Brewster JT, Cook A, Crooks AL, Cruz C, Dwulet NC, Gaudino JJ, Golec D, Harrison JA, Hartley DP, Hassanien SH, Hicken EJ, Kahn D, Laird ER, Lemieux C, Lewandowski N, McCown J, McDonald MG, McNulty O, Mou TC, Nguyen P, Oko L, Opie LP, Otten J, Peck SC, Polites VC, Randall SD, Rosen RZ, Savechenkov P, Simpson H, Singh A, Sparks D, Wickersham K, Wollenberg L, Wong CE, Wong J, Wu WI, Elsayed MSA, Hinklin RJ, Tang TP. Discovery of Pyrazolopyrazines as Selective, Potent, and Mutant-Active MET Inhibitors with Intracranial Efficacy. J Med Chem 2024; 67:14466-14477. [PMID: 39088797 DOI: 10.1021/acs.jmedchem.4c01232] [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: 08/03/2024]
Abstract
Mesenchymal-epithelial transition factor (MET) is a receptor tyrosine kinase that serves a critical function in numerous developmental, morphogenic, and proliferative signaling pathways. If dysregulated, MET has been shown to be involved in the development and survival of several cancers, including non-small cell lung cancer (NSCLC), renal cancer, and other epithelial tumors. Currently, the clinical efficacy of FDA approved MET inhibitors is limited by on-target acquired resistance, dose-limiting toxicities, and less than optimal efficacy against brain metastasis. Therefore, there is still an unmet medical need for the development of MET inhibitors to address these issues. Herein we report the application of structure-based design for the discovery and development of a novel class of brain-penetrant MET inhibitors with enhanced activity against clinically relevant mutations and improved selectivity. Compound 13 with a MET D1228N cell line IC50 value of 23 nM showed good efficacy in an intracranial tumor model and increased the median overall survival of the animals to 100% when dosed orally at 100 mg/kg daily for 21 days.
Collapse
Affiliation(s)
- Quinn A Bumpers
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Robert W Pipal
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Anna M Benz-Weeden
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - James T Brewster
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Adam Cook
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Amy L Crooks
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Cole Cruz
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Natalie C Dwulet
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - John J Gaudino
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Daniel Golec
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Jacqueline A Harrison
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Dylan P Hartley
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Sherif H Hassanien
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Erik J Hicken
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Dean Kahn
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Ellen R Laird
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Christine Lemieux
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Nicholas Lewandowski
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Joseph McCown
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Matthew G McDonald
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Oren McNulty
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Tung-Chung Mou
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Phong Nguyen
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Lauren Oko
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Lisa Pieti Opie
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Jennifer Otten
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Spencer C Peck
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Viktor C Polites
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Samuel D Randall
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Rachel Z Rosen
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Pavel Savechenkov
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Helen Simpson
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Anurag Singh
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Drew Sparks
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Kyle Wickersham
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Lance Wollenberg
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Christina E Wong
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Jim Wong
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Wen-I Wu
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Mohamed S A Elsayed
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Ronald J Hinklin
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| | - Tony P Tang
- Pfizer Research & Development 3200 Walnut Street, Boulder, Colorado 80301, United States
| |
Collapse
|
14
|
Akbar W, Ehsan S, Siddique SA, Sarfraz M, Shaheen F, Shafqat A, Shahnaz, Siddique MBA, Saeed A, Al-Salahi R, El Bakri Y. Solid Phase Synthesis, DFT Calculations, Molecular Docking, and Biological Studies of Symmetrical N 2, N 4, N 6-Trisubstituted-1,3,5-triazines. ACS OMEGA 2024; 9:34428-34444. [PMID: 39157158 PMCID: PMC11325405 DOI: 10.1021/acsomega.4c01980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/26/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024]
Abstract
A diversity-oriented, multicomponent convergent synthesis of symmetrical triazines through a one-pot protocol is presented in this research project. The assembly of trisubstituted triazines was initially carried out using easily available reagents through three different protocols, i.e., conventional, MW-assisted synthesis, and solid-supported MW-assisted synthesis using organic and inorganic support to carry out a comparative analysis as to which procedure best corresponds to a greener synthesis protocol. The compounds formed were characterized for structure elucidation and subjected to in vitro anticancer and antibacterial screening. Additionally, computational studies, such as DFT calculations and molecular docking analyses, were conducted.
Collapse
Affiliation(s)
- Wajiha Akbar
- Department
of Chemistry, Lahore College for Women University, Lahore 44444, Pakistan
| | - Shahana Ehsan
- Department
of Chemistry, Lahore College for Women University, Lahore 44444, Pakistan
| | - Sabir Ali Siddique
- Institute
of Chemistry, The Islamia University of
Bahawalpur, Baghdad-Ul-Jadeed
Campus, Bahawalpur 63100, Pakistan
| | - Muhammad Sarfraz
- Institute
of Chemistry, The Islamia University of
Bahawalpur, Baghdad-Ul-Jadeed
Campus, Bahawalpur 63100, Pakistan
| | - Faiqa Shaheen
- School
of Chemistry, University of the Punjab, Lahore 54590, Pakistan
| | - Ayesha Shafqat
- School
of Botany, Minhaj University, Lahore 54770, Pakistan
| | - Shahnaz
- Department
of Chemistry, Lahore College for Women University, Lahore 44444, Pakistan
| | | | - Ayesha Saeed
- Department
of Chemistry, Lahore College for Women University, Lahore 44444, Pakistan
| | - Rashad Al-Salahi
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Youness El Bakri
- Department
of Theoretical and Applied Chemistry, South
Ural State University, Lenin prospect 76, Chelyabinsk, 454080, Russian Federation
| |
Collapse
|
15
|
Davis JC, Waltz SE. The MET Family of Receptor Tyrosine Kinases Promotes a Shift to Pro-Tumor Metabolism. Genes (Basel) 2024; 15:953. [PMID: 39062731 PMCID: PMC11275592 DOI: 10.3390/genes15070953] [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: 06/26/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
The development and growth of cancer is fundamentally dependent on pro-tumor changes in metabolism. Cancer cells generally shift away from oxidative phosphorylation as the primary source of energy and rely more heavily on glycolysis. Receptor tyrosine kinases (RTKs) are a type of receptor that is implicated in this shift to pro-tumor metabolism. RTKs are important drivers of cancer growth and metastasis. One such family of RTKs is the MET family, which consists of MET and RON (MST1R). The overexpression of either MET or RON has been associated with worse cancer patient prognosis in a variety of tumor types. Both MET and RON signaling promote increased glycolysis by upregulating the expression of key glycolytic enzymes via increased MYC transcription factor activity. Additionally, both MET and RON signaling promote increased cholesterol biosynthesis downstream of glycolysis by upregulating the expression of SREBP2-induced cholesterol biosynthesis enzymes via CTTNB1. These changes in metabolism, driven by RTK activity, provide potential targets in limiting tumor growth and metastasis via pharmacological inhibition or modifications in diet. This review summarizes pro-tumor changes in metabolism driven by the MET family of RTKs. In doing so, we will offer our unique perspective on metabolic pathways that drive worse patient prognosis and provide suggestions for future study.
Collapse
Affiliation(s)
- James C. Davis
- Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Susan E. Waltz
- Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45220, USA
| |
Collapse
|
16
|
Seenan V, Hsu CF, Subramani K, Chen PC, Ding DC, Chu TY. Ovulation provides excessive coagulation and hepatocyte growth factor signals to cause postoperative intraabdominal adhesions. iScience 2024; 27:109788. [PMID: 38770140 PMCID: PMC11103365 DOI: 10.1016/j.isci.2024.109788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/14/2024] [Accepted: 04/16/2024] [Indexed: 05/22/2024] Open
Abstract
Postoperative adhesions show a higher occurrence in females aged 16-60, especially after pelvic surgeries. This study explores the role of ovulation in adhesion formation in mice. Ovarian surgery in mice with normal- or super-ovulation led to pronounced adhesions, whereas ovulation-defective Pgr-KO mice showed minimal adhesions. Specifically, exposure to ovulatory follicular fluid (FF) markedly increased the adhesion. The hazardous exposure time window was one day before to 2.5 days after the surgery. Mechanistically, early FF exposure triggered adhesions via the blood coagulation cascade, while later exposure relied on the HGF/cMET signaling pathway. Prophylactic administration of a thrombin inhibitor pre-operatively or a cMET inhibitor postoperatively effectively mitigated FF-induced adhesions, while COX inhibitor treatment exhibited no discernible effect. These findings underscore ovulation as a pivotal factor in the development of pelvic wound adhesions and advocate for targeted preventive strategies such as c-MET inhibition, scheduling surgeries outside the ovulatory period, or employing oral contraceptive measures.
Collapse
Affiliation(s)
- Vaishnavi Seenan
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan, ROC
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan, ROC
| | - Che-Fang Hsu
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan, ROC
| | - Kanchana Subramani
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan, ROC
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan, ROC
| | - Pao-Chu Chen
- Department of Obstetrics & Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan, ROC
| | - Dah-Ching Ding
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan, ROC
- Department of Obstetrics & Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan, ROC
| | - Tang-Yuan Chu
- Center for Prevention and Therapy of Gynecological Cancers, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan, ROC
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan, ROC
- Department of Obstetrics & Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan, ROC
- Department of Life Sciences, Tzu Chi University, Hualien 970, Taiwan, ROC
| |
Collapse
|
17
|
Zhang P, Xu J, Wu Q, Qian J, Wang S. Development of crizotinib-associated renal cyst in a non-small cell lung cancer patient with ALK fusion: a case report and review of the literature. Diagn Pathol 2024; 19:58. [PMID: 38616252 PMCID: PMC11016210 DOI: 10.1186/s13000-024-01480-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 03/21/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Crizotinib, an oral first-generation tyrosine kinase inhibitor (TKI), is superior to systemic chemotherapy for the treatment of non-small cell lung cancer (NSCLC) with positive rearrangement of anaplastic lymphoma kinase (ALK). However, an increased incidence of renal and hepatic cysts has been reported in the patients on crizotinib treatment. CASE PRESENTATION Here, we describe a case of a 71-year-old Chinese women developed multiple cystic lesions in kidney and liver during crizotinib treatment for the primary and metastatic NSCLC. The renal and hepatic cysts were noted by CT scan 3 months after crizotinib treatment, which were spontaneously and significantly regressed after stopping crizotinib. CONCLUSIONS Based on literature review and our experience in this case report, we concluded that crizotinib-associated renal cyst (CARCs) has features of malignancy and abscess in radiographic imaging, and thus, pathological confirmation is necessary to avoid inappropriate treatment decision. In addition, to benefit the patients with progress-free survival (PFS), switching from crizotinib to alectinib is recommended for the treatment of NSCLC patients who developed CARCs.
Collapse
Affiliation(s)
- Peng Zhang
- Six Departments of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - JiaHua Xu
- Seven Departments of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing Wu
- Seven Departments of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianxin Qian
- Seven Departments of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Song Wang
- Department of Radiology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
18
|
Min W, Wang Y, Shen H, Zheng M, Tong C, Shen H, Wang D, Zhu Y, Wang X, Xiao Y, Zhang XY, Yang P. Discovery of potent and selective c-Met inhibitors for MET-amplified hepatocellular carcinoma treatment. Eur J Med Chem 2024; 264:116025. [PMID: 38086189 DOI: 10.1016/j.ejmech.2023.116025] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023]
Abstract
Hepatocellular carcinoma (HCC) is a prevalent and lethal malignancy worldwide. The MET gene, which encodes receptor tyrosine kinase c-Met, is aberrantly activated in various solid tumors, including non-small cell lung cancer and HCC. In this study, we identified a novel c-Met inhibitor 54 by virtual screening and structural optimization. Compound 54 showed potent c-Met inhibition with an IC50 value of 0.45 ± 0.06 nM. It also exhibited high selectivity among 370 kinases and potent anti-proliferative activity against MET-amplified HCC cells. Moreover, compound 54 displayed significant anti-tumor efficacy in vivo, making it a potential candidate for HCC treatment in future studies.
Collapse
Affiliation(s)
- Wenjian Min
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Yanyin Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Hongtao Shen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Mingming Zheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Chen Tong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Hao Shen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Dawei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Yasheng Zhu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiao Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Yibei Xiao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Xiao-Yu Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China.
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
19
|
de Moraes FCA, Vilbert M, Alves VFC, de Oliveira Almeida G, Priantti JN, Madeira T, Stecca C, Fernandes MR, dos Santos NPC. Mesenchymal-Epithelial Transition Kinase Inhibitor Therapy in Patients with Advanced Papillary Renal-Cell Carcinoma: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:17582. [PMID: 38139411 PMCID: PMC10744118 DOI: 10.3390/ijms242417582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Papillary subtypes of renal-cell carcinoma (pRCC) represent 10-15% of the cases and commonly have MET alterations. This systematic review and single-arm meta-analysis evaluated MET inhibitor therapy (METi) efficacy and safety in adults with confirmed advanced pRCC. The search strategy included PubMed, Web-of-science, Cochrane, and Scopus. We used the DerSimonian/Laird random effect model for all analyses; p-value < 5% was considered significant, and heterogeneity was assessed with I2. Three clinical trials and six cohort studies were included with 504 patients; 31% were MET-driven. Our pooled analysis demonstrated an objective response rate (ORR) in MET-driven, MET-independent, and overall patients of: 36% (95%CI: 10-62), 0% (95%CI: 0-3), and 21% (95%CI: 1-41), respectively. One-year disease control and progression-free survival rates were, respectively, 70% (95%CI: 52-88) and 15% (95%CI: 10-20). Twelve- and twenty-four-month survival rates were, respectively, 43% (95%CI: 23-64) and 10% (95%CI: 0-30). The prevalence of adverse events of any grade and grades 3-5 were 96% (95%CI: 91-100) and 44% (95%CI: 37-50), respectively. We suggest METi has anti-tumor activity and is tolerable in patients with advanced pRCC.
Collapse
Affiliation(s)
| | - Maysa Vilbert
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5T 2S8, Canada
| | | | | | - Jonathan N. Priantti
- School of Medicine, Federal University of Amazonas—UFAM, Manaus 69020-160, Brazil
| | - Thiago Madeira
- School of Medicine, Federal University of Minas Gerais—UFMG, Belo Horizonte 31270-901, Brazil
| | - Carlos Stecca
- Mackenzie Evangelical University Hospital, Curitiba 80710-390, Brazil
| | | | | |
Collapse
|
20
|
Ma H, Wei W, Liang D, Xu X, Yang D, Wang Q, Wang Y, Wei Q, Sun B, Zhao X. HGF-Based CAR-T Cells Target Hepatocellular Carcinoma Cells That Express High Levels of c-Met. Immunol Invest 2023; 52:735-748. [PMID: 37409941 DOI: 10.1080/08820139.2023.2232402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
BACKGROUND CAR-T is emerging as an effective treatment strategy for hematologic malignancies, however its effectiveness for treating solid tumors, such as Hepatocellular Carcinoma (HCC) is limited. Here, we screened a variety of CAR-T cells that target c-Met to investigate their potential to induce HCC cell death in vitro. METHODS Human T cells were transduced to express CARs by lentiviral vector transfection. c-Met expression in human HCC cell lines and CARs expression were monitored by flow cytometry. Tumor cell killing was evaluated by Luciferase Assay System Kit. The concentrations of cytokine were tested by Enzyme-linked immunosorbent assays. Knock down and overexpression studies targeting c-Met were conducted to assess the targeting specificity of CARs. RESULTS We found that CAR T cells expressing a minimal amino-terminal polypeptide sequence comprising the first kringle (kringle 1) domain (denoted as NK1 CAR-T cells), efficiently killed HCC cell lines that expressed high levels of the HGF receptor c-Met. Furthermore, we report that while NK1 CAR-T cells were efficient at targeting SMMC7221 cells for destruction, and its potency was significantly attenuated in parallel experiments with cells stably expressing short hairpin RNAs (shRNAs) that suppressed c-Met expression. Correspondingly, overexpression of c-Met in the embryonic kidney cell line HEK293T led to their enhanced killing by NK1 CAR-T cells. CONCLUSION Our studies demonstrate that a minimal amino-terminal polypeptide sequence comprising the kirngle1 domain of HGF is highly relevant to the design of effective CAR-T cell therapies that kill HCC cells expressing high levels of c-Met.
Collapse
Affiliation(s)
- Haiyan Ma
- Department of Rehabilitation Medicine and Laboratory of Animal Tumor Models, National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenwen Wei
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dandan Liang
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xing Xu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dong Yang
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiong Wang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yun Wang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Quan Wei
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Bin Sun
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xudong Zhao
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Wang J, Tan J, Wu B, Wu R, Han Y, Wang C, Gao Z, Jiang D, Xia X. Customizing cancer treatment at the nanoscale: a focus on anaplastic thyroid cancer therapy. J Nanobiotechnology 2023; 21:374. [PMID: 37833748 PMCID: PMC10571362 DOI: 10.1186/s12951-023-02094-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 09/01/2023] [Indexed: 10/15/2023] Open
Abstract
Anaplastic thyroid cancer (ATC) is a rare but highly aggressive kind of thyroid cancer. Various therapeutic methods have been considered for the treatment of ATC, but its prognosis remains poor. With the advent of the nanomedicine era, the use of nanotechnology has been introduced in the treatment of various cancers and has shown great potential and broad prospects in ATC treatment. The current review meticulously describes and summarizes the research progress of various nanomedicine-based therapeutic methods of ATC, including chemotherapy, differentiation therapy, radioiodine therapy, gene therapy, targeted therapy, photothermal therapy, and combination therapy. Furthermore, potential future challenges and opportunities for the currently developed nanomedicines for ATC treatment are discussed. As far as we know, there are few reviews focusing on the nanomedicine of ATC therapy, and it is believed that this review will generate widespread interest from researchers in a variety of fields to further expedite preclinical research and clinical translation of ATC nanomedicines.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, 430022, Wuhan, Hubei, PR China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, China
| | - Jie Tan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bian Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ruolin Wu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, 430022, Wuhan, Hubei, PR China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, China
| | - Yanmei Han
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, 430022, Wuhan, Hubei, PR China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, China
| | - Chenyang Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, 430022, Wuhan, Hubei, PR China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, China
| | - Zairong Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, 430022, Wuhan, Hubei, PR China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, China.
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, 430022, Wuhan, Hubei, PR China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, China.
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, 430022, Wuhan, Hubei, PR China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, China.
| |
Collapse
|
22
|
Kastana P, Ntenekou D, Mourkogianni E, Enake MK, Xanthopoulos A, Choleva E, Marazioti A, Nikou S, Akwii RG, Papadaki E, Gramage E, Herradón G, Stathopoulos GT, Mikelis CM, Papadimitriou E. Genetic deletion or tyrosine phosphatase inhibition of PTPRZ1 activates c-Met to up-regulate angiogenesis and lung adenocarcinoma growth. Int J Cancer 2023; 153:1051-1066. [PMID: 37260355 PMCID: PMC10524925 DOI: 10.1002/ijc.34564] [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: 11/23/2022] [Revised: 04/12/2023] [Accepted: 04/28/2023] [Indexed: 06/02/2023]
Abstract
Protein tyrosine phosphatase receptor zeta 1 (PTPRZ1) is a transmembrane tyrosine phosphatase (TP) expressed in endothelial cells and required for stimulation of cell migration by vascular endothelial growth factor A165 (VEGFA165 ) and pleiotrophin (PTN). It is also over or under-expressed in various tumor types. In this study, we used genetically engineered Ptprz1-/- and Ptprz1+/+ mice to study mechanistic aspects of PTPRZ1 involvement in angiogenesis and investigate its role in lung adenocarcinoma (LUAD) growth. Ptprz1-/- lung microvascular endothelial cells (LMVEC) have increased angiogenic features compared with Ptprz1+/+ LMVEC, in line with the increased lung angiogenesis and the enhanced chemically induced LUAD growth in Ptprz1-/- compared with Ptprz1+/+ mice. In LUAD cells isolated from the lungs of urethane-treated mice, PTPRZ1 TP inhibition also enhanced proliferation and migration. Expression of beta 3 (β3 ) integrin is decreased in Ptprz1-/- LMVEC, linked to enhanced VEGF receptor 2 (VEGFR2), c-Met tyrosine kinase (TK) and Akt kinase activities. However, only c-Met and Akt seem responsible for the enhanced endothelial cell activation in vitro and LUAD growth and angiogenesis in vivo in Ptprz1-/- mice. A selective PTPRZ1 TP inhibitor, VEGFA165 and PTN also activate c-Met and Akt in a PTPRZ1-dependent manner in endothelial cells, and their stimulatory effects are abolished by the c-Met TK inhibitor (TKI) crizotinib. Altogether, our data suggest that low PTPRZ1 expression is linked to worse LUAD prognosis and response to c-Met TKIs and uncover for the first time the role of PTPRZ1 in mediating c-Met activation by VEGFA and PTN.
Collapse
Affiliation(s)
- Pinelopi Kastana
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | - Despoina Ntenekou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | - Eleni Mourkogianni
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | - Michaela-Karina Enake
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | | | - Effrosyni Choleva
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | - Antonia Marazioti
- Laboratory of Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Greece
| | - Sophia Nikou
- Department of Anatomy, Faculty of Medicine, University of Patras, Greece
| | - Racheal G. Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Centre, Amarillo, TX, USA
| | - Eleni Papadaki
- Department of Anatomy, Faculty of Medicine, University of Patras, Greece
| | - Esther Gramage
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Gonzalo Herradón
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Georgios T. Stathopoulos
- Laboratory of Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Greece
| | - Constantinos M. Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Centre, Amarillo, TX, USA
| | | |
Collapse
|
23
|
Nitulescu GM, Stancov G, Seremet OC, Nitulescu G, Mihai DP, Duta-Bratu CG, Barbuceanu SF, Olaru OT. The Importance of the Pyrazole Scaffold in the Design of Protein Kinases Inhibitors as Targeted Anticancer Therapies. Molecules 2023; 28:5359. [PMID: 37513232 PMCID: PMC10385367 DOI: 10.3390/molecules28145359] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The altered activation or overexpression of protein kinases (PKs) is a major subject of research in oncology and their inhibition using small molecules, protein kinases inhibitors (PKI) is the best available option for the cure of cancer. The pyrazole ring is extensively employed in the field of medicinal chemistry and drug development strategies, playing a vital role as a fundamental framework in the structure of various PKIs. This scaffold holds major importance and is considered a privileged structure based on its synthetic accessibility, drug-like properties, and its versatile bioisosteric replacement function. It has proven to play a key role in many PKI, such as the inhibitors of Akt, Aurora kinases, MAPK, B-raf, JAK, Bcr-Abl, c-Met, PDGFR, FGFRT, and RET. Of the 74 small molecule PKI approved by the US FDA, 8 contain a pyrazole ring: Avapritinib, Asciminib, Crizotinib, Encorafenib, Erdafitinib, Pralsetinib, Pirtobrutinib, and Ruxolitinib. The focus of this review is on the importance of the unfused pyrazole ring within the clinically tested PKI and on the additional required elements of their chemical structures. Related important pyrazole fused scaffolds like indazole, pyrrolo[1,2-b]pyrazole, pyrazolo[4,3-b]pyridine, pyrazolo[1,5-a]pyrimidine, or pyrazolo[3,4-d]pyrimidine are beyond the subject of this work.
Collapse
Affiliation(s)
| | | | | | - Georgiana Nitulescu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (G.M.N.)
| | | | | | | | | |
Collapse
|
24
|
Das B, Sethy C, Chatterjee S, Dash SR, Sinha S, Paul S, Goutam K, Kundu CN. Quinacrine inhibits cMET-mediated metastasis and angiogenesis in breast cancer stem cells. J Cell Commun Signal 2023:10.1007/s12079-023-00756-9. [PMID: 37162635 DOI: 10.1007/s12079-023-00756-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/24/2023] [Indexed: 05/11/2023] Open
Abstract
A trans-membrane receptor tyrosine kinase, cMET, belonging to the MET proto-oncogene family, is responsible for cancer metastasis and angiogenesis. But not much is known about the role of cMET in growth and progression of cancer stem cells (CSCs). Earlier studies have shown that Quinacrine (QC), a bioactive agent, has anti-CSCs activity. Here, the role of QC in deregulation of cMET-mediated metastasis and angiogenesis has been systematically evaluated in vitro in highly metastatic breast CSCs (mBCSCs), ex vivo in patient-derived breast cancer stem cells (PDBCSCs) and in vivo in xenograft mice model systems. Cell proliferation, migration, invasion and representative metastasis markers were upregulated in cMET-overexpressed cells and QC exposure inhibited these processes in both mBCSCs and PDBCSCs. Interestingly, metastasis was significantly inhibited by QC in cMET-overexpressed cells but comparatively lesser significant alteration of the process was noted in cMET-silenced cells. Increase in vascularization (in in ovo CAM assay), and cell-cell tube formation (in HUVECs), and enhanced MMP9 and MMP2 enzymatic activities (in gelatin zymography) were noted after cMET overexpression but these processes got reversed after cMET knockdown or QC treatment in cMET-overexpressed cells. QC inhibited angiogenesis significantly in cMET-overexpressed cells, but lesser significant change was observed in cMET-silenced cells. Reduction in tumor volume and decreased expression of metastatic and angiogenic markers were also noted in xenograft mice after QC treatment. Furthermore, QC inhibited cMET activity by dephosphorylation of its tyrosine residues (Y1234 and Y1356) and downregulation of its downstream cascade. Thus, QC inhibited the cMET-mediated metastasis and angiogenesis in in vitro, in ovo, in vivo and ex vivo model systems. Ligand (HGF) binding leads to receptor dimerization and phosphorylation of tyrosine kinase domain of cMET. This activates the cMET signaling cascade. The representative downstream metastasis and angiogenesis-related proteins get upregulated and induce the metastasis and angiogenesis process. But after the QC treatment, cMET get dephosphorylated and inactivated. As a result, the downstream signaling proteins of cMET along with the other representative metastatic and angiogenic factors get downregulated. These lead to inhibition of cMET-mediated metastasis and angiogenesis. (Created with BioRender.com).
Collapse
Affiliation(s)
- Biswajit Das
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Chinmayee Sethy
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Subhajit Chatterjee
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Somya Ranjan Dash
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Saptarshi Sinha
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Subarno Paul
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Kunal Goutam
- Department of Surgical Oncology, Acharya Harihar Regional Cancer Centre, Cuttack, Odisha, 753007, India
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
25
|
Bhoopathi P, Mannangatti P, Das SK, Fisher PB, Emdad L. Chemoresistance in pancreatic ductal adenocarcinoma: Overcoming resistance to therapy. Adv Cancer Res 2023; 159:285-341. [PMID: 37268399 DOI: 10.1016/bs.acr.2023.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a prominent cause of cancer deaths worldwide, is a highly aggressive cancer most frequently detected at an advanced stage that limits treatment options to systemic chemotherapy, which has provided only marginal positive clinical outcomes. More than 90% of patients with PDAC die within a year of being diagnosed. PDAC is increasing at a rate of 0.5-1.0% per year, and it is expected to be the second leading cause of cancer-related mortality by 2030. The resistance of tumor cells to chemotherapeutic drugs, which can be innate or acquired, is the primary factor contributing to the ineffectiveness of cancer treatments. Although many PDAC patients initially responds to standard of care (SOC) drugs they soon develop resistance caused partly by the substantial cellular heterogeneity seen in PDAC tissue and the tumor microenvironment (TME), which are considered key factors contributing to resistance to therapy. A deeper understanding of molecular mechanisms involved in PDAC progression and metastasis development, and the interplay of the TME in all these processes is essential to better comprehend the etiology and pathobiology of chemoresistance observed in PDAC. Recent research has recognized new therapeutic targets ushering in the development of innovative combinatorial therapies as well as enhancing our comprehension of several different cell death pathways. These approaches facilitate the lowering of the therapeutic threshold; however, the possibility of subsequent resistance development still remains a key issue and concern. Discoveries, that can target PDAC resistance, either alone or in combination, have the potential to serve as the foundation for future treatments that are effective without posing undue health risks. In this chapter, we discuss potential causes of PDAC chemoresistance and approaches for combating chemoresistance by targeting different pathways and different cellular functions associated with and mediating resistance.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
26
|
Liu C, Zhang Y, Sun W, Zhu H, Su M, Wang X, Rong X, Wang K, Yu M, Sheng W, Zhu B. A novel GSH-activable theranostic probe containing kinase inhibitor for synergistic treatment and selective imaging of tumor cells. Talanta 2023; 260:124567. [PMID: 37121140 DOI: 10.1016/j.talanta.2023.124567] [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: 01/14/2023] [Revised: 04/05/2023] [Accepted: 04/15/2023] [Indexed: 05/02/2023]
Abstract
Theranostic probe is becoming a powerful tool for diagnosis and treatment of cancer. Although some theranostic probes have been successfully developed, there is still a great room for improvement in sensitive diagnosis and efficient treatment. Herein, we developed a novel GSH-activable theranostic probe NC-G, which uses 1,8-naphthalimide-4-sulfonamide as a fluorescence imaging group and crizotinib as a highly toxic kinase inhibitor to tumor cells. The probe not only has high sensitivity (DL = 74 nM) and specificity, but also can detect GSH sensitively in cells and zebrafish. In addition, probe NC-G can not only show more obvious fluorescence in tumor cells to achieve sensitive diagnosis of tumor cells, but also release the inhibitor crizotinib to achieve high toxicity to tumor cells. It is worth noting that the consumption of GSH can cause oxidative stress response of cells and the release of SO2 can induce cell apoptosis during the recognition process of the probe and GSH. Thus, the synergistic effect of crizotinib, GSH depletion, and SO2 release provides a highly effective therapeutic feature for tumor cells. Therefore, probe NC-G can serve as an excellent theranostic probe for sensitive imaging and highly effective treatment of tumor cells.
Collapse
Affiliation(s)
- Caiyun Liu
- School of Water Conservancy and Environment, University of Jinan, Jinan, 250022, China.
| | - Yan Zhang
- School of Water Conservancy and Environment, University of Jinan, Jinan, 250022, China
| | - Weimin Sun
- School of Water Conservancy and Environment, University of Jinan, Jinan, 250022, China
| | - Hanchuang Zhu
- School of Water Conservancy and Environment, University of Jinan, Jinan, 250022, China
| | - Meijun Su
- School of Water Conservancy and Environment, University of Jinan, Jinan, 250022, China
| | - Xin Wang
- School of Water Conservancy and Environment, University of Jinan, Jinan, 250022, China
| | - Xiaodi Rong
- School of Water Conservancy and Environment, University of Jinan, Jinan, 250022, China
| | - Kun Wang
- School of Water Conservancy and Environment, University of Jinan, Jinan, 250022, China
| | - Miaohui Yu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China
| | - Wenlong Sheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250103, China.
| | - Baocun Zhu
- School of Water Conservancy and Environment, University of Jinan, Jinan, 250022, China.
| |
Collapse
|
27
|
Huang YC, Hsieh PY, Wang LY, Tsai TH, Chen YJ, Hsieh CH. Local Liver Irradiation Concurrently Versus Sequentially with Cabozantinib on the Pharmacokinetics and Biodistribution in Rats. Int J Mol Sci 2023; 24:ijms24065849. [PMID: 36982920 PMCID: PMC10056485 DOI: 10.3390/ijms24065849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 03/22/2023] Open
Abstract
The aim of this study was to evaluate the radiotherapy (RT)-pharmacokinetics (PK) effect of cabozantinib in concurrent or sequential regimens with external beam radiotherapy (EBRT) or stereotactic body radiation therapy (SBRT). Concurrent and sequential regimens involving RT and cabozantinib were designed. The RT–drug interactions of cabozantinib under RT were confirmed in a free-moving rat model. The drugs were separated on an Agilent ZORBAX SB-phenyl column with a mobile phase consisting of 10 mM potassium dihydrogen phosphate (KH2PO4)–methanol solution (27:73, v/v) for cabozantinib. There were no statistically significant differences in the concentration versus time curve of cabozantinib (AUCcabozantinib) between the control group and the RT2Gy×3 f’x and RT9Gy×3 f’x groups in the concurrent and the sequential regimens. However, compared to those in the control group, the Tmax, T1/2 and MRT decreased by 72.8% (p = 0.04), 49.0% (p = 0.04) and 48.5% (p = 0.04) with RT2Gy×3 f’x in the concurrent regimen, respectively. Additionally, the T1/2 and MRT decreased by 58.8% (p = 0.01) and 57.8% (p = 0.01) in the concurrent RT9Gy×3 f’x group when compared with the control group, respectively. The biodistribution of cabozantinib in the heart increased by 271.4% (p = 0.04) and 120.0% (p = 0.04) with RT2Gy×3 f’x in the concurrent and sequential regimens compared to the concurrent regimen, respectively. Additionally, the biodistribution of cabozantinib in the heart increased by 107.1% (p = 0.01) with the RT9Gy×3 f’x sequential regimen. Compared to the RT9Gy×3 f’x concurrent regimen, the RT9Gy×3 f’x sequential regimen increased the biodistribution of cabozantinib in the heart (81.3%, p = 0.02), liver (110.5%, p = 0.02), lung (125%, p = 0.004) and kidneys (87.5%, p = 0.048). No cabozantinib was detected in the brain in any of the groups. The AUC of cabozantinib is not modulated by irradiation and is not affected by treatment strategies. However, the biodistribution of cabozantinib in the heart is modulated by off-target irradiation and SBRT doses simultaneously. The impact of the biodistribution of cabozantinib with RT9Gy×3 f’x is more significant with the sequential regimen than with the concurrent regimen.
Collapse
Affiliation(s)
- Yu-Chuen Huang
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan (Y.-J.C.)
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan
| | - Pei-Ying Hsieh
- Department of Oncology and Hematology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Li-Ying Wang
- School and Graduate Institute of Physical Therapy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Physical Therapy Center, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Yu-Jen Chen
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan (Y.-J.C.)
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Department of Radiation Oncology, Mackay Memorial Hospital, Taipei 104, Taiwan
- Department of Artificial Intelligence and Medical Application, MacKay Junior College of Medicine, Nursing, and Management, Taipei 112, Taiwan
| | - Chen-Hsi Hsieh
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Correspondence:
| |
Collapse
|
28
|
Parejo-Alonso B, Royo-García A, Espiau-Romera P, Courtois S, Curiel-García Á, Zagorac S, Villaoslada I, Olive KP, Heeschen C, Sancho P. Pharmacological targeting of the receptor ALK inhibits tumorigenicity and overcomes chemoresistance in pancreatic ductal adenocarcinoma. Biomed Pharmacother 2023; 158:114162. [PMID: 36571997 DOI: 10.1016/j.biopha.2022.114162] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive disease characterized by its metastatic potential and chemoresistance. These traits are partially attributable to the highly tumorigenic pancreatic cancer stem cells (PaCSCs). Interestingly, these cells show unique features in order to sustain their identity and functionality, some of them amenable for therapeutic intervention. Screening of phospho-receptor tyrosine kinases revealed that PaCSCs harbored increased activation of anaplastic lymphoma kinase (ALK). We subsequently demonstrated that oncogenic ALK signaling contributes to tumorigenicity in PDAC patient-derived xenografts (PDXs) by promoting stemness through ligand-dependent activation. Indeed, the ALK ligands midkine (MDK) or pleiotrophin (PTN) increased self-renewal, clonogenicity and CSC frequency in several in vitro local and metastatic PDX models. Conversely, treatment with the clinically-approved ALK inhibitors Crizotinib and Ensartinib decreased PaCSC content and functionality in vitro and in vivo, by inducing cell death. Strikingly, ALK inhibitors sensitized chemoresistant PaCSCs to Gemcitabine, as the most used chemotherapeutic agent for PDAC treatment. Consequently, ALK inhibition delayed tumor relapse after chemotherapy in vivo by effectively decreasing the content of PaCSCs. In summary, our results demonstrate that targeting the MDK/PTN-ALK axis with clinically-approved inhibitors impairs in vivo tumorigenicity and chemoresistance in PDAC suggesting a new treatment approach to improve the long-term survival of PDAC patients.
Collapse
Affiliation(s)
- Beatriz Parejo-Alonso
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Alba Royo-García
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Pilar Espiau-Romera
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Sarah Courtois
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Álvaro Curiel-García
- Department of Medicine, Division of Digestive Liver Diseases and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Sladjana Zagorac
- Center for Stem Cells in Cancer & Ageing (Barts Cancer Institute), London, UK
| | - Isabel Villaoslada
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Hospital Universitario Miguel Servet, Zaragoza, Spain; Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Kenneth P Olive
- Department of Medicine, Division of Digestive Liver Diseases and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Christopher Heeschen
- Center for Single-Cell Omics and Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, China; Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute - FPO - IRCCS, Candiolo (Torino), Italy
| | - Patricia Sancho
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Hospital Universitario Miguel Servet, Zaragoza, Spain.
| |
Collapse
|
29
|
Cecchi F, Rex K, Schmidt J, Vocke CD, Lee YH, Burkett S, Baker D, Damore MA, Coxon A, Burgess TL, Bottaro DP. Rilotumumab Resistance Acquired by Intracrine Hepatocyte Growth Factor Signaling. Cancers (Basel) 2023; 15:460. [PMID: 36672409 PMCID: PMC9857108 DOI: 10.3390/cancers15020460] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Drug resistance is a long-standing impediment to effective systemic cancer therapy and acquired drug resistance is a growing problem for molecularly-targeted therapeutics that otherwise have shown unprecedented successes in disease control. The hepatocyte growth factor (HGF)/Met receptor pathway signaling is frequently involved in cancer and has been a subject of targeted drug development for nearly 30 years. To anticipate and study specific resistance mechanisms associated with targeting this pathway, we engineered resistance to the HGF-neutralizing antibody rilotumumab in glioblastoma cells harboring autocrine HGF/Met signaling, a frequent abnormality of this brain cancer in humans. We found that rilotumumab resistance was acquired through an unusual mechanism comprising dramatic HGF overproduction and misfolding, endoplasmic reticulum (ER) stress-response signaling and redirected vesicular trafficking that effectively sequestered rilotumumab and misfolded HGF from native HGF and activated Met. Amplification of MET and HGF genes, with evidence of rapidly acquired intron-less, reverse-transcribed copies in DNA, was also observed. These changes enabled persistent Met pathway activation and improved cell survival under stress conditions. Point mutations in the HGF pathway or other complementary or downstream growth regulatory cascades that are frequently associated with targeted drug resistance in other prevalent cancer types were not observed. Although resistant cells were significantly more malignant, they retained sensitivity to Met kinase inhibition and acquired sensitivity to inhibition of ER stress signaling and cholesterol biosynthesis. Defining this mechanism reveals details of a rapidly acquired yet highly-orchestrated multisystem route of resistance to a selective molecularly-targeted agent and suggests strategies for early detection and effective intervention.
Collapse
Affiliation(s)
- Fabiola Cecchi
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karen Rex
- Amgen, Inc., Thousand Oaks, CA 91320, USA
| | | | - Cathy D. Vocke
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Young H. Lee
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sandra Burkett
- Molecular Cytogenetics Core Facility, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | | | | | | | | | - Donald P. Bottaro
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
30
|
Oh SY, Lee YW, Lee EJ, Kim JH, Park Y, Heo SG, Yu MR, Hong MH, DaSilva J, Daly C, Cho BC, Lim SM, Yun MR. Preclinical Study of a Biparatopic METxMET Antibody-Drug Conjugate, REGN5093-M114, Overcomes MET-driven Acquired Resistance to EGFR TKIs in EGFR-mutant NSCLC. Clin Cancer Res 2023; 29:221-232. [PMID: 36269795 DOI: 10.1158/1078-0432.ccr-22-2180] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/08/2022] [Accepted: 10/19/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE MET amplification is a frequent mechanism of resistance to EGFR tyrosine kinase inhibitors (TKI) in patients with EGFR-mutated non-small cell lung cancer (NSCLC), and combined treatment with EGFR TKIs and MET TKIs has been explored as a strategy to overcome resistance. However, durable response is invariably limited by the emergence of acquired resistance. Here, we investigated the preclinical activity of REGN5093-M114, a novel antibody-drug conjugate targeting MET in MET-driven patient-derived models. EXPERIMENTAL DESIGN Patient-derived organoids, patient-derived cells, or ATCC cell lines were used to investigate the in vitro/in vivo activity of REGN5093-M114. RESULTS REGN5093-M114 exhibited significant antitumor efficacy compared with MET TKI or unconjugated METxMET biparatopic antibody (REGN5093). Regardless of MET gene copy number, MET-overexpressed TKI-naïve EGFR-mutant NSCLC cells responded to REGN5093-M114 treatment. Cell surface MET expression had the most predictive power in determining the efficacy of REGN5093-M114. REGN5093-M114 potently reduced tumor growth of EGFR-mutant NSCLC with PTEN loss or MET Y1230C mutation after progression on prior osimertinib and savolitinib treatment. CONCLUSIONS Altogether, REGN5093-M114 is a promising candidate to overcome the challenges facing functional MET pathway blockade.
Collapse
Affiliation(s)
- Seung Yeon Oh
- Department of Research Support, Yonsei Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
- Department of Biomedical Science institute, Graduated School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - You Won Lee
- Department of Research Support, Yonsei Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Eun Ji Lee
- Department of Research Support, Yonsei Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
- Department of Biomedical Science institute, Graduated School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Jae Hwan Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - YoungJoon Park
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Seong Gu Heo
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Mi Ra Yu
- Department of Research Support, Yonsei Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Min Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - John DaSilva
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of South Korea
| | - Mi Ran Yun
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of South Korea
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of South Korea
| |
Collapse
|
31
|
[Research Progresses in the Treatment of NSCLC with MET Gene Variants: A Riview]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:877-887. [PMID: 36617474 PMCID: PMC9845091 DOI: 10.3779/j.issn.1009-3419.2022.101.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesenchymal-epithelial transition factor (MET) has long been considered as the most crucial and promising driver gene in the occurrence and development of non-small cell lung cancer (NSCLC), except for epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), and c-ROS oncogene 1 receptor tyrosine kinase (ROS1). In recent years, therapeutic drugs targeting MET have been continuously developed and applied in clinical practice. First, the curative effect of NSCLC patients with MET exon 14 skipping mutations has been further improved. In addition, when MET amplification occurs after resistance to EGFR tyrosine kinase inhibitors (EGFR-TKIs) in patients with advanced EGFR-mutant NSCLC, the combination of MET-TKIs and EGFR-TKIs has brought significant survival benefits and many other advances. This article reviews the treatment progress of NSCLC patients with different types of MET variants under different circumstances, which provides reference for the selection of clinical treatment strategies.
.
Collapse
|
32
|
Sounbuli K, Mironova N, Alekseeva L. Diverse Neutrophil Functions in Cancer and Promising Neutrophil-Based Cancer Therapies. Int J Mol Sci 2022; 23:ijms232415827. [PMID: 36555469 PMCID: PMC9779721 DOI: 10.3390/ijms232415827] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Neutrophils represent the most abundant cell type of leukocytes in the human blood and have been considered a vital player in the innate immune system and the first line of defense against invading pathogens. Recently, several studies showed that neutrophils play an active role in the immune response during cancer development. They exhibited both pro-oncogenic and anti-tumor activities under the influence of various mediators in the tumor microenvironment. Neutrophils can be divided into several subpopulations, thus contradicting the traditional concept of neutrophils as a homogeneous population with a specific function in the innate immunity and opening new horizons for cancer therapy. Despite the promising achievements in this field, a full understanding of tumor-neutrophil interplay is currently lacking. In this review, we try to summarize the current view on neutrophil heterogeneity in cancer, discuss the different communication pathways between tumors and neutrophils, and focus on the implementation of these new findings to develop promising neutrophil-based cancer therapies.
Collapse
Affiliation(s)
- Khetam Sounbuli
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
- Faculty of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Nadezhda Mironova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
- Correspondence: ; Tel.: +7-383-363-51-61
| | - Ludmila Alekseeva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentiev Ave., 8, Novosibirsk 630090, Russia
| |
Collapse
|
33
|
Ma YX, Liu FR, Zhang Y, Chen Q, Chen ZQ, Liu QW, Huang Y, Yang YP, Fang WF, Xi N, Kang N, Zhuang YL, Zhang Q, Jiang YZ, Zhang L, Zhao HY. Preclinical characterization and phase I clinical trial of CT053PTSA targets MET, AXL, and VEGFR2 in patients with advanced solid tumors. Front Immunol 2022; 13:1024755. [PMID: 36341335 PMCID: PMC9632963 DOI: 10.3389/fimmu.2022.1024755] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/30/2022] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND CT053PTSA is a novel tyrosine kinase inhibitor that targets MET, AXL, VEGFR2, FLT3 and MERTK. Here, we present preclinical data about CT053PTSA, and we conducted the first-in-human (FIH) study to evaluate the use of CT053PTSA in adult patients with pretreated advanced solid tumors. METHODS The selectivity and antitumor activity of CT053PTSA were assessed in cell lines in vitro through kinase and cellular screening panels and in cell line-derived tumor xenograft (CDX) and patient-derived xenograft (PDX) models in vivo. The FIH, phase I, single-center, single-arm, dose escalation (3 + 3 design) study was conducted, patients received at least one dose of CT053PTSA (15 mg QD, 30 mg QD, 60 mg QD, 100 mg QD, and 150 mg QD). The primary objectives were to assess safety and tolerability, to determine the maximum tolerated dose (MTD), dose-limiting toxicity (DLT), and the recommended dose of CT053PTSA for further study. Secondary objectives included pharmacokinetics, antitumor activity. RESULTS CT053 (free-base form of CT053PTSA) inhibited MET, AXL, VEGFR2, FLT3 and MERTK phosphorylation and suppressed tumor cell angiogenesis by blocking VEGF and HGF, respectively, in vitro. Moreover, cell lines with high MET expression exhibited strong sensitivity to CT053, and CT053 blocked the MET and AXL signaling pathways. In an in vivo study, CT053 significantly inhibited tumor growth in CDX and PDX models. Twenty eligible patients were enrolled in the FIH phase I trial. The most common treatment-related adverse events were transaminase elevation (65%), leukopenia (45%) and neutropenia (35%). DLTs occurred in 3 patients, 1/6 in the 100 mg group and 2/4 in the 150 mg group, so the MTD was set to 100 mg. CT053PTSA was rapidly absorbed after the oral administration of a single dose, and the Cmax and AUC increased proportionally as the dose increased. A total of 17 patients in this trial underwent tumor imaging evaluation, and 29.4% had stable disease. CONCLUSIONS CT053PTSA has potent antitumor and antiangiogenic activity in preclinical models. In this FIH phase I trial, CT053PTSA was well tolerated and had a satisfactory safety profile. Further trials evaluating the clinical activity of CT053PTSA are ongoing.
Collapse
Affiliation(s)
- Yu-Xiang Ma
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Fu-Rong Liu
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yang Zhang
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qun Chen
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhi-Qiang Chen
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qian-Wen Liu
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yan Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yun-Peng Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wen-Feng Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ning Xi
- HEC R&D Center, Sunshine Lake Pharma Co., Ltd, Donggguan, China
| | - Ning Kang
- HEC R&D Center, Sunshine Lake Pharma Co., Ltd, Donggguan, China
| | - Yu-Lei Zhuang
- HEC R&D Center, Sunshine Lake Pharma Co., Ltd, Donggguan, China
| | - Qi Zhang
- HEC R&D Center, Sunshine Lake Pharma Co., Ltd, Donggguan, China
| | - Ying-Zhi Jiang
- HEC R&D Center, Sunshine Lake Pharma Co., Ltd, Donggguan, China
| | - Li Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hong-Yun Zhao
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
34
|
Management of Peripheral Edema in Patients with MET Exon 14-Mutated Non-small Cell Lung Cancer Treated with Small Molecule MET Inhibitors. Target Oncol 2022; 17:597-604. [PMID: 36087188 PMCID: PMC9512730 DOI: 10.1007/s11523-022-00912-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2022] [Indexed: 11/03/2022]
Abstract
Small molecule mesenchymal-epithelial transition (MET) inhibitors, such as crizotinib, capmatinib, and tepotinib, are treatment options for metastatic non-small cell lung cancer (NSCLC) in adult patients whose tumors have a mutation that leads to MET exon 14 skipping. In clinical trials, these MET inhibitors were associated with a high incidence of peripheral edema, although this was generally mild-to-moderate in severity. There is limited information about the mechanism involved in MET inhibitor-induced peripheral edema. Perturbation of hepatocyte growth factor (HGF)/MET signaling may disrupt the permeability balance in the vascular endothelium and thus promote edema development. Another potential mechanism is through effects on renal function, although this is unlikely to be the primary mechanism. Because edema is common in cancer patients and may not necessarily be caused by the cancer treatment, or other conditions that have similar symptoms to peripheral edema, a thorough assessment is required to ascertain the underlying cause. Before starting MET-inhibitor therapy, patients should be educated about the possibility of developing peripheral edema. Patient limb volume should be measured before initiating treatment, to aid assessment if symptoms develop. Since the exact mechanism of MET inhibitor-induced edema is unknown, management is empiric, with common approaches including compression stockings, specific exercises, massage, limb elevation, and/or diuretic treatment. Although not usually required, discontinuation of MET inhibitor treatment generally resolves peripheral edema. Early diagnosis and management, as well as patient information and education, are vital to decrease the clinical burden associated with edema, and to reinforce capmatinib treatment adherence.
Collapse
|
35
|
Yao H, Ren Y, Yan J, Liu J, Hu J, Yan M, Li X. Design, Synthesis, and Evaluation of New Mesenchymal–Epithelial Transition Factor (c-Met) Kinase Inhibitors with Dual Chiral Centers. Molecules 2022; 27:molecules27175359. [PMID: 36080127 PMCID: PMC9457593 DOI: 10.3390/molecules27175359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
A series of tepotinib derivatives with two chiral centers was designed, synthesized, and evaluated as anticancer agents. The optimal compound (R, S)-12a strongly exhibited antiproliferative activity against MHCC97H cell lines with an IC50 value of 0.002 μM, compared to tepotinib (IC50 = 0.013 μM). Mechanistic studies revealed that compound (R, S)-12a significantly inhibited c-Met activation, as well as the downstream AKT signaling pathway, and suppressed wound closure. Moreover, compound (R, S)-12a induced cellular apoptosis and cell cycle arrest at the G1 phase in a dose-dependent fashion.
Collapse
Affiliation(s)
- Han Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuanyuan Ren
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jun Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiadai Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jinhui Hu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
- Correspondence: (J.H.); (M.Y.)
| | - Ming Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Correspondence: (J.H.); (M.Y.)
| | - Xingshu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
36
|
Collie GW, Barlind L, Bazzaz S, Börjesson U, Dale IL, Disch JS, Habeshian S, Jetson R, Khurana P, Madin A, Michaelides IN, Peng L, Snijder A, Stubbs CJ. Discovery of a selective c-MET inhibitor with a novel binding mode. Bioorg Med Chem Lett 2022; 75:128948. [PMID: 35987508 DOI: 10.1016/j.bmcl.2022.128948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/30/2022]
Abstract
The c-MET receptor tyrosine kinase has received considerable attention as a cancer drug target yet there remains a need for inhibitors which are selective for c-MET and able to target emerging drug-resistant mutants. We report here the discovery, by screening a DNA-encoded chemical library, of a highly selective c-MET inhibitor which was shown by X-ray crystallography to bind to the kinase in an unprecedented manner. These results represent a novel mode of inhibiting c-MET with a small molecule and may provide a route to targeting drug-resistant forms of the kinase whilst avoiding potential toxicity issues associated with broad kinome inhibition.
Collapse
Affiliation(s)
| | - Louise Barlind
- Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Sana Bazzaz
- X-Chem, Inc., Waltham, MA 02453, United States
| | - Ulf Börjesson
- Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Ian L Dale
- Discovery Sciences, R&D, AstraZeneca, Cambridge, U.K
| | | | | | | | | | - Andrew Madin
- Discovery Sciences, R&D, AstraZeneca, Cambridge, U.K
| | | | - Ling Peng
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, PR China
| | - Arjan Snijder
- Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | |
Collapse
|
37
|
Hepatocyte Growth Factor Enhances Antineoplastic Effect of 5-Fluorouracil by Increasing UPP1 Expression in HepG2 Cells. Int J Mol Sci 2022; 23:ijms23169108. [PMID: 36012373 PMCID: PMC9409026 DOI: 10.3390/ijms23169108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Aberrant activation of hepatocyte growth factor (HGF) and its receptor c-Met axis promotes tumor growth. Therefore, many clinical trials have been conducted. A phase 3 trial investigating a monoclonal antibody targeting HGF in combination with fluoropyrimidine-based chemotherapy had to be terminated prematurely; however, the reason behind the failure remains poorly defined. In this study, we investigated the influence of HGF on the antineoplastic effects of 5-fluorouracil (5-FU), a fluoropyrimidine, in HepG2 cells. HGF suppressed the proliferative activity of cells concomitantly treated with 5-FU more robustly as compared to that of cells treated with 5-FU alone, and markedly increased the expression of uridine phosphorylase 1 (UPP1). Intracellular concentration of 5-fluorouridine, an initial anabolite of 5-FU catalyzed by UPP1, was increased by HGF. Interestingly, erlotinib enhanced HGF-induced increase in UPP1 mRNA; in contrast, gefitinib suppressed it. Furthermore, erlotinib suppressed HGF-increased phosphorylation of the epidermal growth factor receptor at the Tyr1173 site involved in downregulation of extracellular signal-regulated kinase (Erk) activation, and enhanced the HGF-increased phosphorylation of Erk. Collectively, these findings suggest that inhibition of the HGF/c-Met axis diminishes the effects of fluoropyrimidine through downregulation of UPP1 expression. Therefore, extreme caution must be exercised in terms of patient safety while offering chemotherapy comprising fluoropyrimidine concomitantly with inhibitors of the HGF/c-Met axis.
Collapse
|
38
|
Singh AK, Malviya R. Coagulation and inflammation in cancer: Limitations and prospects for treatment. Biochim Biophys Acta Rev Cancer 2022; 1877:188727. [PMID: 35378243 DOI: 10.1016/j.bbcan.2022.188727] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 02/08/2023]
Abstract
The development of so-called immune checkpoint inhibitors (ICIs), which target specific molecular processes of tumour growth, has had a transformative effect on cancer treatment. Widespread use of antibody-based medicines to inhibit tumour cell immune evasion by modulating T cell responses is becoming more common. Despite this, response rates are still low, and secondary resistance is an issue that arises often. In addition, a wide range of serious adverse effects is triggered by enhancing the immunological response. As a result of an increased mortality rate, a higher prevalence of thrombotic complications is connected with an increased incidence of immunological reactions, complement activation, and skin toxicity. This suggests that the tumour microenvironment's interaction between coagulation and inflammation is important at every stage of the tumour's life cycle. The coagulation system's function in tumour formation is the topic of this review. By better understanding the molecular mechanisms in which tumour cells circulate, plasmatic coagulation and immune system cells are engaged, new therapy options for cancer sufferers may be discovered.
Collapse
Affiliation(s)
- Arun Kumar Singh
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
39
|
Wang Y, Liu T, Chen G, Gong J, Bai Y, Zhang T, Xu N, Liu L, Xu J, He J, Liu Y, Zhang L, Jiang D, Wang M, Chang J, Li W, Bai C, Zhou J, Wang J, Ren Y, Zhang L, Su W, Liu, B, Shen L. Phase Ia/Ib Study of the Selective MET Inhibitor, Savolitinib, in Patients with Advanced Solid Tumors:
Safety, Efficacy, and Biomarkers. Oncologist 2022; 27:oyab066. [PMID: 35445725 PMCID: PMC9074963 DOI: 10.1093/oncolo/oyab066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Savolitinib has shown good tolerability and preliminary efficacy, but efficacy biomarkers require investigation. The main purpose of this study was to confirm in Chinese patients the recommended phase II dose (RP2D) of savolitinib and to explore overall benefit in tumors bearing c-Met aberration. METHODS This was an open-label, multi-center, 2-part phase I study. A starting dose of 600 mg QD was initiated in the escalation phase, utilizing a 3+3 design with repeated QD and BID dosing. In the dose expansion phase, we enrolled patients with gastric cancer and non-small cell lung cancer (NSCLC) with documented c-met aberration into 5 cohorts to further explore biomarkers. c-Met overexpression and amplification were assessed by immunohistochemistry and FISH, respectively. RESULTS The safety analysis set included 85 patients. Only one dose-limiting toxicity (grade 3 fatigue) was reported in the 600 mg BID dosing group. The most frequent treatment-related adverse events were nausea (29.4%), vomiting (27.1%), and peripheral edema (21.2%). Notably, in gastric cancer, response was only observed in patients with MET amplification (copy number 9.7-18.4), with an objective response rate of 35.7% and a disease control rate of 64.3%. For patients with NSCLC bearing a MET exon 14 skipping mutation, obvious target lesion shrinkage was observed in 2 of 4 patients, although PR was not achieved. CONCLUSION The RP2D of savolitinib was established as 600 mg QD or 500 mg BID in Chinese patients. The promising response observed in patients with gastric cancer with c-met amplification and NSCLC with MET exon 14 skipping mutation warrants further investigation. CLINICALTRIALS.GOV IDENTIFIER NCT0198555.
Collapse
Affiliation(s)
- Yakun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, and Center of Evidence-Based Medicine, Fudan University, Shanghai, People’s Republic of China
| | - Gongyan Chen
- Department of Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Jifang Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology/Early Drug Development Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Yuxian Bai
- Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, People’s Republic of China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jianming Xu
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Jianxing He
- Department of Thoracic Surgery/Oncology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
- State Key Laboratory and National Clinical Research Center for Respiratory Disease, Guangzhou, People’s Republic of China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, People’s Republic of China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Da Jiang
- Department of Medical Oncology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Jianhua Chang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
- Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, People’s Republic of China
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Chunmei Bai
- Department of Medical Oncology, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Beijing, People’s Republic of China
| | - Jinghong Zhou
- Hutchison China MediTech Limited, Shanghai, People’s Republic of China
| | - Jian Wang
- Hutchison China MediTech Limited, Shanghai, People’s Republic of China
| | - Yongxin Ren
- Hutchison China MediTech Limited, Shanghai, People’s Republic of China
| | - Liya Zhang
- Hutchison China MediTech Limited, Shanghai, People’s Republic of China
| | - Weiguo Su
- Hutchison China MediTech Limited, Shanghai, People’s Republic of China
| | - Baorui Liu,
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, People’s Republic of China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology/Early Drug Development Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| |
Collapse
|
40
|
Hamilton G, Rath B. Met inhibitors in the treatment of lung cancer: the evidence to date. Expert Opin Pharmacother 2022; 23:815-825. [PMID: 35377279 DOI: 10.1080/14656566.2022.2062227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : The hepatocyte growth factor (HGF) receptor MET is an oncogenic driver in a subpopulation of Non-small Lung Cancer Cells (NSCLC) at the primary tumor stage or in acquired resistance to treatment with tumor-targeting tyrosine kinase inhibitors (TKIs). AREAS COVERED This article summarizes the mechanisms leading to overexpression and activation of MET by amplification and mutations including exon 14 aberrations. Furthermore, the methods to detect and categorize MET as a tumor driver and the selective TKIs for patient treatment are discussed. EXPERT OPINION : Activating mutations and rearrangements of kinases in NSCLC are the target of successful therapeutic intervention. However, MET activation involves a number of complex alterations including gene amplification, prevention of degradation by METex14 exon skipping and a host of gene mutations. A high-level of MET expression is the precondition for tumor responses to TKIs and the confirmation of MET-dependent tumor progression is difficult in primary lesions and in tumors exhibiting resistance to mutated EGFR-directed therapy in absence of standardized and concordant assays of MET amplification.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Department of Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
Zhang J, Dai J, Lan X, Zhao Y, Yang F, Zhang H, Tang S, Liang G, Wang X, Tang Q. Synthesis, bioevaluation and molecular dynamics of pyrrolo-pyridine benzamide derivatives as potential antitumor agents in vitro and in vivo. Eur J Med Chem 2022; 233:114215. [DOI: 10.1016/j.ejmech.2022.114215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/17/2022] [Accepted: 02/19/2022] [Indexed: 11/04/2022]
|
42
|
Sochacka-Ćwikła A, Mączyński M, Regiec A. FDA-Approved Small Molecule Compounds as Drugs for Solid Cancers from Early 2011 to the End of 2021. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072259. [PMID: 35408658 PMCID: PMC9000317 DOI: 10.3390/molecules27072259] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 01/09/2023]
Abstract
Solid cancers are the most common types of cancers diagnosed globally and comprise a large number of deaths each year. The main challenge currently in drug development for tumors raised from solid organs is to find more selective compounds, which exploit specific molecular targets. In this work, the small molecule drugs registered by the Food and Drug Administration (FDA) for solid cancers treatment between 2011 and 2022 were identified and analyzed by investigating a type of therapy they are used for, as well as their structures and mechanisms of action. On average, 4 new small molecule agents were introduced each year, with a few exceptions, for a total of 62 new drug approvals. A total of 50 of all FDA-approved drugs have also been authorized for use in the European Union by the European Medicines Agency (EMA). Our analysis indicates that many more anticancer molecules show a selective mode of action, i.e., 49 targeted agents, 5 hormone therapies and 3 radiopharmaceuticals, compared to less specific cytostatic action, i.e., 5 chemotherapeutic agents. It should be emphasized that new medications are indicated for use mainly for monotherapy and less for a combination or adjuvant therapies. The comprehensive data presented in this review can serve for further design and development of more specific targeted agents in clinical usage for solid tumors.
Collapse
|
43
|
Bukhari SNA. Synthesis and evaluation of new chalcones and oximes as anticancer agents. RSC Adv 2022; 12:10307-10320. [PMID: 35424971 PMCID: PMC8973297 DOI: 10.1039/d2ra01198k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/28/2022] [Indexed: 12/28/2022] Open
Abstract
Complex illnesses, such as cancer, are often caused by many disorders, gene mutations, or pathways. Biological pathways play a significant part in the development of these diseases. Multi-target directed ligands (MTDLs) have been used by medicinal chemists recently in an effort to find single molecules that can affect many targets concurrently. In this work, several chalcones containing the ligustrazine moiety were synthesized and tested for their in vitro anticancer activity and several cancer markers, including EGFR, BRAFV600E, c-Met, and tubulin polymerization, in order to uncover multitarget bioactive compounds. In assays using multiple cancer cell lines, the majority of the compounds examined showed strong anticancer activity against them. To synthesize oximes, all of the chalcones were used as precursors. The IC50 values of two compounds (11g and 11e) were found to be 0.87, 0.28, 2.43, 1.04 μM and 11d, 1.47, 0.79, 3.8, 1.63 μM respectively, against A-375, MCF-7, HT-29 and H-460 cell lines. These IC50 values revealed an excellent antiproliferative activity compared to those of the positive control foretinib, (IC50 = 1.9, 1.15, 3.97, and 2.86 μM). Careful examination of their structure and configuration revealed that both compounds had an oxime functional group with z configuration, in place of carbonyl functional group, along with a 2-phenyl thiophenyl moiety with or without a bromo group at position-5. The possible binding pattern was implied by docking simulation, inferring the possibility of introducing interactions with the nearby tubulin chain. Since the novel structural trial has been conducted with a detailed structure activity relationship discussion, this work might stimulate new ideas in further modification of multitarget anti-cancer agents and therapeutic approaches. Discovery of multitarget anticancer agents by modifications of natural compound.![]()
Collapse
Affiliation(s)
- Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University Sakaka Aljouf 72388 Saudi Arabia +96 6565738896
| |
Collapse
|
44
|
Dong Y, Xu J, Sun B, Wang J, Wang Z. MET-Targeted Therapies and Clinical Outcomes: A Systematic Literature Review. Mol Diagn Ther 2022; 26:203-227. [PMID: 35266116 PMCID: PMC8942886 DOI: 10.1007/s40291-021-00568-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2021] [Indexed: 12/17/2022]
Abstract
Introduction Numerous therapeutic agents specifically targeting the mesenchymal-epithelial transition (MET) oncogene are being developed. Objective The aim of the current review was to systematically identify and analyze clinical trials that have evaluated MET inhibitors in various cancer types and to provide an overview of their clinical outcomes. Methods An electronic literature search was carried out in the PubMed and Embase databases to identify published clinical trials related to MET inhibitors. The PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) statement was followed for the systematic appraisal of the literature. Data related to clinical outcomes, including progression-free survival, overall survival, objective response rate, and overall tumor response, were extracted. Results In total, 49 publications were included. Among these, 51.02% were phase II studies, 14.28% were randomized controlled trials, three were phase III studies, two were prospective observational studies, and the remainder were either phase I or Ib studies. The majority (44.89%) of articles reported the clinical outcomes of MET inhibitors, including small molecules, monoclonal antibodies, and other agents, in patients with non-small-cell lung cancer (NSCLC) harboring MET alterations. MET amplification, overexpression, and MET exon 14 skipping mutations were the major MET alteration types reported across the included studies. Clinical responses/outcomes varied considerably. Conclusion This systematic literature review provides an overview of the literature available in Embase and PubMed regarding MET-targeted therapies. MET-selective tyrosine kinase inhibitors (TKIs) (capmatinib, tepotinib, and savolitinib) may become a new standard of care in NSCLC, specifically with MET exon 14 skipping mutations. A combination of MET TKIs with epidermal growth factor receptor (EGFR) TKIs (osimertinib + savolitinib, tepotinib + gefitinib) may be a potential solution for MET-driven EGFR TKI resistance. Further, MET alteration (MET amplification/overexpression) may be an actionable target in gastric cancer and papillary renal cell carcinoma.
Collapse
Affiliation(s)
- Yiting Dong
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China
| | - Jiachen Xu
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China
| | - Boyang Sun
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China.
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
45
|
Mead EA, Boulghassoul-Pietrzykowska N, Wang Y, Anees O, Kinstlinger NS, Lee M, Hamza S, Feng Y, Pietrzykowski AZ. Non-Invasive microRNA Profiling in Saliva can Serve as a Biomarker of Alcohol Exposure and Its Effects in Humans. Front Genet 2022; 12:804222. [PMID: 35126468 PMCID: PMC8812725 DOI: 10.3389/fgene.2021.804222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Alcohol Use Disorder (AUD) is one of the most prevalent mental disorders worldwide. Considering the widespread occurrence of AUD, a reliable, cheap, non-invasive biomarker of alcohol consumption is desired by healthcare providers, clinicians, researchers, public health and criminal justice officials. microRNAs could serve as such biomarkers. They are easily detectable in saliva, which can be sampled from individuals in a non-invasive manner. Moreover, microRNAs expression is dynamically regulated by environmental factors, including alcohol. Since excessive alcohol consumption is a hallmark of alcohol abuse, we have profiled microRNA expression in the saliva of chronic, heavy alcohol abusers using microRNA microarrays. We observed significant changes in salivary microRNA expression caused by excessive alcohol consumption. These changes fell into three categories: downregulated microRNAs, upregulated microRNAs, and microRNAs upregulated de novo. Analysis of these combinatorial changes in microRNA expression suggests dysregulation of specific biological pathways leading to impairment of the immune system and development of several types of epithelial cancer. Moreover, some of the altered microRNAs are also modulators of inflammation, suggesting their contribution to pro-inflammatory mechanisms of alcohol actions. Establishment of the cellular source of microRNAs in saliva corroborated these results. We determined that most of the microRNAs in saliva come from two types of cells: leukocytes involved in immune responses and inflammation, and buccal cells, involved in development of epithelial, oral cancers. In summary, we propose that microRNA profiling in saliva can be a useful, non-invasive biomarker allowing the monitoring of alcohol abuse, as well as alcohol-related inflammation and early detection of cancer.
Collapse
Affiliation(s)
- Edward A. Mead
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nadia Boulghassoul-Pietrzykowska
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Mayo Clinic Health System, NWWI, Barron, WI, United States
- Department of Medicine, Capital Health, Trenton, NJ, United States
- Weight and Life MD, Hamilton, NJ, United States
| | - Yongping Wang
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Holmdel Township School, Holmdel, NJ, United States
| | - Onaiza Anees
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Virginia Commonwealth University Health, CMH Behavioral Health, South Hill, VA, United States
| | - Noah S. Kinstlinger
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maximillian Lee
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- George Washington University, School of Medicine and Health Sciences, Washington DC, MA, United States
| | - Shireen Hamza
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Department of the History of Science, Harvard University, Cambridge, MA, United States
| | - Yaping Feng
- Waksman Genomics Core Facility, Rutgers University, Piscataway, NJ, United States
- Bioinformatics Department, Admera Health, South Plainfield, NJ, United States
| | - Andrzej Z. Pietrzykowski
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Weight and Life MD, Hamilton, NJ, United States
| |
Collapse
|
46
|
O'Sullivan Coyne G, Kummar S, Hu J, Ganjoo K, Chow WA, Do KT, Zlott J, Bruns A, Rubinstein L, Foster JC, Juwara L, Meehan R, Piekarz R, Streicher H, Sharon E, Takebe N, Voth AR, Bottaro D, Costello R, Wright JJ, Doroshow JH, Chen AP. Clinical Activity of Single-Agent Cabozantinib (XL184), a Multi-receptor Tyrosine Kinase Inhibitor, in Patients with Refractory Soft-Tissue Sarcomas. Clin Cancer Res 2022; 28:279-288. [PMID: 34716194 PMCID: PMC8776602 DOI: 10.1158/1078-0432.ccr-21-2480] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/17/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Soft-tissue sarcomas (STS) are a rare, heterogeneous group of mesenchymal tumors. For decades the mainstay of treatment for advanced, unresectable STS has been palliative chemotherapy. High levels of activated MET receptor have been reported in various sarcoma cell lines, together with elevated vascular endothelial growth factor (VEGF) levels in patients with STS, suggesting that dual targeting of the VEGF and MET pathways with the multi-receptor tyrosine kinase inhibitor cabozantinib would result in clinical benefit in this population. PATIENTS AND METHODS We performed an open-label, multi-institution, single-arm phase II trial of single-agent cabozantinib in adult patients with advanced STS and progressive disease after at least 1 standard line of systemic therapy. Patients received 60 mg oral cabozantinib once daily in 28-day cycles, and dual primary endpoints of overall response rate and 6-month progression-free survival (PFS) were assessed. Changes in several circulating biomarkers were assessed as secondary endpoints. RESULTS Six (11.1%; 95% CI, 4.2%-22.6%) of the 54 evaluable patients enrolled experienced objective responses (all partial responses). Six-month PFS was 49.3% (95% CI, 36.2%-67.3%), with a median time on study of 4 cycles (range, 1-99). The most common grade 3/4 adverse events were hypertension (7.4%) and neutropenia (16.7%). Patients' levels of circulating hepatocyte growth factor (HGF), soluble MET, and VEGF-A generally increased after a cycle of therapy, while soluble VEGFR2 levels decreased, regardless of clinical outcome. CONCLUSIONS Cabozantinib single-agent antitumor activity was observed in patients with selected STS histologic subtypes (alveolar soft-part sarcoma, undifferentiated pleomorphic sarcoma, extraskeletal myxoid chondrosarcoma, and leiomyosarcoma) highlighting the biomolecular diversity of STS.
Collapse
Affiliation(s)
- Geraldine O'Sullivan Coyne
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Shivaani Kummar
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - James Hu
- University of Southern California, Los Angeles, California
| | - Kristen Ganjoo
- Stanford Cancer Center, Stanford University, Palo Alto, California
| | | | - Khanh T Do
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Jennifer Zlott
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Ashley Bruns
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Lawrence Rubinstein
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Jared C Foster
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Lamin Juwara
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Robert Meehan
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Richard Piekarz
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Howard Streicher
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Elad Sharon
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Naoko Takebe
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Andrea Regier Voth
- Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Donald Bottaro
- Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Rene Costello
- Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - John J Wright
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - James H Doroshow
- Center for Cancer Research, NCI, NIH, Bethesda, Maryland
- Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Alice P Chen
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland.
| |
Collapse
|
47
|
Fogli S, Tabbò F, Capuano A, Re MD, Passiglia F, Cucchiara F, Scavone C, Gori V, Novello S, Schmidinger M, Danesi R. The expanding family of c-Met inhibitors in solid tumors: a comparative analysis of their pharmacologic and clinical differences. Crit Rev Oncol Hematol 2022; 172:103602. [DOI: 10.1016/j.critrevonc.2022.103602] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 12/16/2022] Open
|
48
|
Bauer AT, Gorzelanny C, Gebhardt C, Pantel K, Schneider SW. Interplay between coagulation and inflammation in cancer: Limitations and therapeutic opportunities. Cancer Treat Rev 2022; 102:102322. [DOI: 10.1016/j.ctrv.2021.102322] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022]
|
49
|
Grundy M, Narendran A. The hepatocyte growth factor/mesenchymal epithelial transition factor axis in high-risk pediatric solid tumors and the anti-tumor activity of targeted therapeutic agents. Front Pediatr 2022; 10:910268. [PMID: 36034555 PMCID: PMC9399617 DOI: 10.3389/fped.2022.910268] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/15/2022] [Indexed: 01/04/2023] Open
Abstract
Clinical trials completed in the last two decades have contributed significantly to the improved overall survival of children with cancer. In spite of these advancements, disease relapse still remains a significant cause of death in this patient population. Often, increasing the intensity of current protocols is not feasible because of cumulative toxicity and development of drug resistance. Therefore, the identification and clinical validation of novel targets in high-risk and refractory childhood malignancies are essential to develop effective new generation treatment protocols. A number of recent studies have shown that the hepatocyte growth factor (HGF) and its receptor Mesenchymal epithelial transition factor (c-MET) influence the growth, survival, angiogenesis, and metastasis of cancer cells. Therefore, the c-MET receptor tyrosine kinase and HGF have been identified as potential targets for cancer therapeutics and recent years have seen a race to synthesize molecules to block their expression and function. In this review we aim to summarize the literature that explores the potential and biological rationale for targeting the HGF/c-MET pathway in common and high-risk pediatric solid tumors. We also discuss selected recent and ongoing clinical trials with these agents in relapsed pediatric tumors that may provide applicable future treatments for these patients.
Collapse
Affiliation(s)
- Megan Grundy
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Aru Narendran
- POETIC Laboratory for Preclinical and Drug Discovery Studies, Division of Pediatric Oncology, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
50
|
Pingaew R, Choomuenwai V, Leechaisit R, Prachayasittikul V, Prachayasittikul S, Prachayasittikul V. 1,2,3-Triazole Scaffold in Recent Medicinal Applications: Synthesis and Anticancer Potentials. HETEROCYCLES 2022. [DOI: 10.3987/rev-22-sr(r)4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|