1
|
Almeida GS, King P, Hallsworth A, Webber H, Popov S, Miranda S, Yogev O, Pearson ADJ, Chesler L, Jamin Y, Robinson SP. Response of GEM models of neuroblastoma to cabozantinib assessed by multiparametric magnetic resonance imaging. Neoplasia 2025; 65:101170. [PMID: 40359728 DOI: 10.1016/j.neo.2025.101170] [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: 09/14/2024] [Accepted: 04/23/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND In neuroblastoma MYCN amplification is associated with enhanced angiogenesis and poor survival. Mutations in the anaplastic lymphoma kinase (ALK) gene can occur with MYCN amplification, conferring a very poor prognosis. Vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF)/c-MET signalling are implicated in neuroblastoma progression. Cabozantinib has potent activity against VEGFR2 and MET. METHODS The efficacy of cabozantinib against tumours arising in GEM models of high-risk neuroblastoma was assessed using multiparametric MRI. Tumour-bearing Th-MYCN and Th-ALKF1174L/Th-MYCN mice were imaged prior to, 24 and 48 hrs after treatment with either 30mg/kg/day cabozantinib or vehicle. Treatment-induced changes in tumour volume, native T1, R2* and ADC were evaluated, and histological correlates sought. Additional Th-MYCN mice were treated daily for up to 28 days. RESULTS Cabozantinib elicited significant 24 and 60 % growth delay 24 and 48 hrs after treatment in tumours in Th-MYCN mice, and a significant 6-8 % reduction in native T1. Tumour R2* was significantly reduced 48 hrs post-treatment. Significantly higher tumour necrosis and apoptosis, and significantly lower Ki67, CD34 and VEGFR2 staining, was determined from the cabozantinib-treated mice. Treatment of Th-ALKF1174L/Th-MYCN mice caused significant 4 % and 21 % tumour growth delay, and a significant 5 % reduction in native T1 at 48 hrs. Daily cabozantinib treatment of Th-MYCN mice elicited significant tumour growth delay over 7 days which translated into significant survival benefit. CONCLUSION Cabozantinib exhibits activity against neuroblastomas arising in both Th-MYCN and Th-MYCN/ALKF1174L mice, revealed in situ using MRI. Native T1 is an early, sensitive and clinically translatable imaging biomarker of effective treatment response in neuroblastoma.
Collapse
Affiliation(s)
- Gilberto S Almeida
- Division of Radiotherapy & Imaging, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom; Division of Clinical Studies, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom
| | - Philippa King
- Division of Clinical Studies, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom
| | - Albert Hallsworth
- Division of Clinical Studies, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom
| | - Hannah Webber
- Division of Clinical Studies, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom
| | - Sergey Popov
- Division of Molecular Pathology, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom
| | - Susana Miranda
- Division of Clinical Studies, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom
| | - Orli Yogev
- Division of Clinical Studies, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom
| | - Andrew D J Pearson
- Division of Clinical Studies, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom
| | - Louis Chesler
- Division of Clinical Studies, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom
| | - Yann Jamin
- Division of Radiotherapy & Imaging, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom
| | - Simon P Robinson
- Division of Radiotherapy & Imaging, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom.
| |
Collapse
|
2
|
Zhang W, Hong X, Xiao Y, Wang H, Zeng X. Sorafenib resistance and therapeutic strategies in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2025; 1880:189310. [PMID: 40187502 DOI: 10.1016/j.bbcan.2025.189310] [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: 11/08/2024] [Revised: 03/30/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most prevalent and lethal cancers globally. While surgical resection and liver transplantation offer potential cures for early-stage HCC, the majority of patients are diagnosed at advanced stages where such interventions are not viable. Sorafenib, a multi-target kinase inhibitor, has been a cornerstone in the treatment of advanced HCC since its approval in 2007. Despite its significant clinical impact, less than half of the treated patients derive long-term benefits due to the emergence of resistance and associated side effects. This review focuses on the role of sorafenib, an FDA-approved multi-target kinase inhibitor, in treating advanced HCC, discusses the mechanisms underlying its therapeutic effects and associated resistance, and explores additional therapeutic strategies being investigated to improve patient outcomes.
Collapse
Affiliation(s)
- Weijing Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Xuechuan Hong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuling Xiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Xiaodong Zeng
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China.
| |
Collapse
|
3
|
Fang B, Lu Y, Li X, Wei Y, Ye D, Wei G, Zhu Y. Targeting the tumor microenvironment, a new therapeutic approach for prostate cancer. Prostate Cancer Prostatic Dis 2025; 28:260-269. [PMID: 38565910 DOI: 10.1038/s41391-024-00825-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND A growing number of studies have shown that in addition to adaptive immune cells such as CD8 + T cells and CD4 + T cells, various other cellular components within prostate cancer (PCa) tumor microenvironment (TME), mainly tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs) and myeloid-derived suppressor cells (MDSCs), have been increasingly recognized as important modulators of tumor progression and promising therapeutic targets. OBJECTIVE In this review, we aim to delineate the mechanisms by which TAMs, CAFs and MDSCs interact with PCa cells in the TME, summarize the therapeutic advancements targeting these cells and discuss potential new therapeutic avenues. METHODS We searched PubMed for relevant studies published through December 10 2023 on TAMs, CAFs and MDSCs in PCa. RESULTS TAMs, CAFs and MDSCs play a critical role in the tumorigenesis, progression, and metastasis of PCa. Moreover, they substantially mediate therapeutic resistance against conventional treatments including anti-androgen therapy, chemotherapy, and immunotherapy. Therapeutic interventions targeting these cellular components have demonstrated promising effects in preclinical models and several clinical trials for PCa, when administrated alone, or combined with other anti-cancer therapies. However, the lack of reliable biomarkers for patient selection and incomplete understanding of the mechanisms underlying the interactions between these cellular components and PCa cells hinder their clinical translation and utility. CONCLUSION New therapeutic strategies targeting TAMs, CAFs, and MDSCs in PCa hold promising prospects. Future research endeavors should focus on a more comprehensive exploration of the specific mechanisms by which these cells contribute to PCa, aiming to identify additional drug targets and conduct more clinical trials to validate the safety and efficacy of these treatment strategies.
Collapse
Affiliation(s)
- Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Gonghong Wei
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| |
Collapse
|
4
|
Xie T, Hu W, You L, Wang X. Design, synthesis and biological evaluation of thienopyridine derivatives as c-Met kinase inhibitors. Mol Divers 2025; 29:2391-2405. [PMID: 39356364 DOI: 10.1007/s11030-024-10998-3] [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: 06/27/2024] [Accepted: 09/22/2024] [Indexed: 10/03/2024]
Abstract
With cabozantinib as the precursor, a novel small molecule inhibitors of c-Met kinase with thieno [2,3-b] pyridine as the scaffold were designed, synthesized and evaluated for their biological activity against A549, Hela and MCF-7 cell lines. The in vitro activities of 16 compounds were tested by MTT method with cabozantinib as control drug. Most compounds had moderate to strong inhibitory activities on cells. Among them, compound 10 had the strongest inhibitory activity, which was superior to the lead compound cabozantinib. Its IC50 values for A549, Hela and MCF-7 cells were 0.005, 2.833 and 13.581 μM, respectively. The colony formation assay demonstrated that compound 10 significantly inhibited the colony formation of A549 cells and suppressed their growth in a concentration-dependent manner. The wound healing assay showed that compound 10 could effectively inhibit the migration of cancer cells compared to a blank control group. The AO/EB assay demonstrated that compound 10 possesses the capability to effectively trigger apoptosis in a concentration-dependent manner. The elementary structure-activity relationship, molecular docking and pharmacokinetics studies revealed the significance of thieno [2,3-b] pyridine derivatives in anti-tumor activity.
Collapse
Affiliation(s)
- Tianyu Xie
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110036, China
| | - Wenbo Hu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110036, China
| | - Lin You
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110036, China
| | - Xin Wang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110036, China.
- Liaoning Key Laboratory of New Drug Research & Development, Shenyang, 110036, China.
| |
Collapse
|
5
|
Maruyama S, Kobayashi H, Hiraga T, Anno T, Sanjo T, Arai M, Ishida M, Kanno H, Kato M. Association of Plasma Cabozantinib Concentration With Treatment Response and Adverse Events in Japanese Patients With Advanced Renal Cell Carcinoma. Ther Drug Monit 2025; 47:400-406. [PMID: 39208400 DOI: 10.1097/ftd.0000000000001254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/25/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Cabozantinib is highly effective against advanced renal cell carcinoma (RCC). However, approximately 60% of the patients require a dose reduction due to severe adverse events. Although associations between trough concentrations of cabozantinib and its efficacy and safety have been reported in other countries, reports on Japanese patients are unavailable. Therefore, we investigated the association of cabozantinib trough concentration with therapeutic efficacy and adverse events in Japanese patients with RCC and evaluated the usefulness of therapeutic drug monitoring. METHODS In this prospective observational study, we measured the trough concentrations of cabozantinib in 10 Japanese patients with RCC enrolled between May 2022 and September 2023. The associations of trough concentration with treatment response, as determined by RECIST 1.1, and the occurrence of grade 2 or higher adverse events were assessed. RESULTS Trough concentration was higher in patients with controlled cancer than in those with progressive cancer (1024 ± 352 versus 457 ± 216 ng/mL, P = 0.035). In addition, patients with grade 2 or higher adverse events showed a significantly higher trough concentration than those without (1560 ± 513 versus 807 ± 319 ng/mL, P = 0.032). In particular, grade 2 or higher dysgeusia, anorexia, fatigue, and dyspepsia significantly correlated with trough concentrations. CONCLUSIONS This is the first clinical study to demonstrate a correlation between cabozantinib trough concentration, therapeutic efficacy, and adverse events in Japanese patients with RCC. The therapeutic drug monitoring of cabozantinib could be useful for improving therapeutic efficacy and avoiding serious adverse events.
Collapse
Affiliation(s)
- Shinichi Maruyama
- Department of Bioanalytical Chemistry, Showa University Graduate School of Pharmacy, Tokyo, Japan
- Departments of Pharmacy and
| | - Hiroaki Kobayashi
- Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
- Department of Urology, National Defense Medical College Hospital, Saitama, Japan
| | | | - Tadatsugu Anno
- Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
| | - Tansei Sanjo
- Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
| | - Masashi Arai
- Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
| | - Masaru Ishida
- Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
| | | | - Masaru Kato
- Department of Bioanalytical Chemistry, Showa University Graduate School of Pharmacy, Tokyo, Japan
| |
Collapse
|
6
|
Xue X, Duan X, Qin M, Liu S, Su L, Cao X, Duan H, Liu B, Ni T, Li X. Topical application of Cap-loaded hydrogels inhibits corneal neovascularization. Exp Eye Res 2025; 255:110390. [PMID: 40246162 DOI: 10.1016/j.exer.2025.110390] [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: 11/20/2024] [Revised: 03/17/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
Corneal neovascularization (CNV) is a significant risk factor for visual impairment. The efficiency and side effects of current CNV treatments, such as steroids and antivascular endothelial growth factor agents, are still debated. In addition, the bioavailability of topical drugs is usually hindered by tears, blinking, and the corneal anatomy. Therefore, finding a new therapeutic strategy is important. This study aimed to examine the function of the new therapeutic agent capmatinib (Cap), a highly selective inhibitor of MET that plays an important role in angiogenesis, in treating CNV. In this study, we first investigated the role of the HGF/c-MET axis in CNV and the therapeutic effect of Cap in a corneal alkali burn model. We synthesised a genipin-crosslinked gelatine-based hydrogel containing Cap (Cap-Gel). We observed a more significant therapeutic effect with the Cap-Gel than with Cap alone, as well as the alleviation of inflammatory infiltration and fibrosis. On day 14, the Cap-Gel group showed the most significant inhibition of corneal neovascularization, with the shortest neovessel length (0.48 ± 0.13 mm), smallest CNV area (3.77 ± 0.78 mm2), and lowest clinical assessment score (3.33 ± 0.52). Taken together, our results suggest that Cap-Gel could be a promising drug candidate for treating CNV.
Collapse
Affiliation(s)
- Xiaoyang Xue
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, PR China
| | - Xiaochuan Duan
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, 300134, PR China
| | - Mengyuan Qin
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China
| | - Shoukuan Liu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, PR China
| | - Lin Su
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, PR China
| | - Xin Cao
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China
| | - Hongquan Duan
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, PR China; Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, PR China; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, 300070, PR China
| | - Boshi Liu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, PR China.
| | - Tianwen Ni
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, PR China.
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, PR China.
| |
Collapse
|
7
|
Yim J, Hope C, Huelse JM, Graham DK. Prospects of current AXL-targeting therapies in early phase cancer trials. Expert Opin Investig Drugs 2025:1-33. [PMID: 40413629 DOI: 10.1080/13543784.2025.2511178] [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: 01/08/2025] [Revised: 04/22/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
INTRODUCTION AXL, a member of the TAM (TYRO3, AXL, and MERTK) family of receptor tyrosine kinases, controls pro-tumorigenic signaling cascades and cancer-immunological functions, and promotes drug resistance. Due to AXL's multifaceted role and therapeutic activity in preclinical studies, a variety of AXL inhibitors are being developed and tested in clinical trials for cancer treatment. Some clinical studies are showing promising results for AXL inhibitors as monotherapy and in combination with standard of care therapeutics. Currently, no selective AXL-targeting therapy has reached FDA-approval, but several compounds have entered phase II and III studies. AREA COVERED We elaborate on the role of AXL in cancer progression and suppressing anti-cancer immunity at both the molecular level and immune cell interaction level. Additionally, we review pre-clinical and clinical data of AXL-targeting agents. EXPERT OPINION Preclinical and several early clinical trials demonstrated the safety of AXL-targeting monotherapies with some evidence of efficacy. Additionally, multiple novel combination regimens including AXL-targeting agents to overcome resistance mechanisms are being actively examined with some promising results. However, patient selection and companion biomarkers may be critical for the success of AXL-targeting therapies.
Collapse
Affiliation(s)
- Juhye Yim
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Chloe Hope
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Justus M Huelse
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University, Atlanta, GA, USA
| |
Collapse
|
8
|
Yang K, Zhang X, Yang K, Liu S, Zhang J, Fu Y, Liu T, Wu K, Li J, Liu C, Huang Q, Qu K. Overexpression of c-Myc triggers p62 aggregation-mediated mitochondrial mitophagy in cabozantinib resistance of hepatocellular carcinoma. Mol Med 2025; 31:209. [PMID: 40426058 PMCID: PMC12107842 DOI: 10.1186/s10020-025-01263-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Resistance to tyrosine kinase inhibitors (TKIs) poses a significant challenge in the treatment of hepatocellular carcinoma (HCC). Although dysregulation of mitochondrial dynamics has been implicated in the aggressive behaviors of various tumors, the specific role and underlying mechanisms by which this dysregulation contributes to cabozantinib resistance in HCC cells remains insufficiently characterized. By investigating mitochondrial dynamics as central regulators of cabozantinib resistance, this work specifically aims to discover actionable targets for restoring drug sensitivity in treatment-refractory HCC cells. We employed transmission electron microscopy (TEM) and confocal microscopy to analyze mitochondrial morphology in HCC cells resistant to TKIs. Additionally, we utilized an oncogene hydrodynamic injection-induced primary liver cancer mouse model to assess the therapeutic efficacy of combining cabozantinib with other pharmacological agents. Our results demonstrated significant increases in mitochondrial fragmentation, p62 aggregation, and mitophagy in cabozantinib-resistant HCC cells, which correlated with overexpression of c-Myc. Notably, inhibiting mitochondrial fission, p62 aggregation, or autophagy effectively reversed the resistance of HCC cells to cabozantinib. Mechanistically, cabozantinib treatment was shown to induce c-Myc expression, which significantly enhanced mitochondrial fragmentation and p62 aggregation, thereby promoting mitophagy. This mitophagic process selectively eliminated damaged mitochondria, reducing cytochrome C-induced apoptosis in cabozantinib-resistant cells. Ultimately, combining cabozantinib with either the autophagy inhibitor chloroquine or the p62 aggregation inhibitor XRK3F2 resulted in improved anticancer efficacy. In conclusion, c-Myc overexpression facilitates p62 aggregation-mediated mitophagy, leading to cabozantinib resistance in HCC cells. Inhibition of autophagy effectively restores cabozantinib sensitivity in HCC.
Collapse
Affiliation(s)
- Kaibo Yang
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China
| | - Xing Zhang
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China
| | - Kun Yang
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China
| | - Sinan Liu
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China
- Department of Surgical Intensive Care Unit, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jingyao Zhang
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China
- Department of Surgical Intensive Care Unit, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yunong Fu
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China
| | - Tong Liu
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China
| | - Kunjin Wu
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China
| | - Jing Li
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China
| | - Chang Liu
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China.
| | - Qichao Huang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Kai Qu
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710004, China.
| |
Collapse
|
9
|
Yao SX, Huang YJ, Zhang YX, Cui ZX, Lu HY, Wang R, Shi L. Revisiting VEGF/VEGFR-2 signalling as an anticancer target and its inhibitor discovery: where are we and where should we go? J Drug Target 2025:1-24. [PMID: 40387416 DOI: 10.1080/1061186x.2025.2508985] [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: 02/04/2025] [Revised: 04/30/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Angiogenesis plays an important role in tumour growth and metastasis. Targeting tumour vascular endothelial cells to inhibit tumour angiogenesis and thus block tumour blood and nutrition supply is the current research focus on anti-tumour growth and metastasis. Vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR-2) signal pathway regulates the proliferation, migration, survival and angiogenesis of vascular endothelial cells, which is abnormally activated in different tumours. Studies have confirmed that inhibiting VEGF/VEGFR-2 signalling pathway can produce anti-tumour effect. Nowadays, anti-angiogenesis therapy targeting VEGF/VEGFR-2 inhibition has become the most effective clinical strategy for cancer treatment. Therefore, a variety of VEGF/VEGFR-2 inhibitors with different structures have been developed. A few selectively inhibit VEGF to block the activation of VEGFR-2 pathway, while the majority selectively inhibit VEGFR-2 as multi-target inhibitors. Based on the classification of dominant skeletons, this paper briefly analyzes the biological activity, clinical research process and structure-activity relationship of the representative small molecule inhibitors of VEGF/VEGFR-2.
Collapse
Affiliation(s)
- Sheng-Xin Yao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yu-Jing Huang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yue-Xi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ze-Xi Cui
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | | | - Ru Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lei Shi
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
10
|
Schweickert PG, Piovesan D, Mitchell CG, Zepeda-Carranza B, Zhu WS, Lopez Espinoza AY, Rocha L, Singh J, Malgapo MIP, Meleza C, Northington KR, Ray RD, Zhao X, Lawson KV, Walters MJ, Sivick KE. Casdatifan (AB521) is a novel and potent allosteric small molecule inhibitor of protumourigenic HIF-2α dependent transcription. Br J Pharmacol 2025. [PMID: 40400177 DOI: 10.1111/bph.70075] [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: 08/23/2024] [Revised: 03/20/2025] [Accepted: 04/18/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND AND PURPOSE Hypoxia-inducible factor 2α (HIF-2α) is a transcription factor that mediates the expression of genes critical for cell adaptation and survival in low oxygen (hypoxic) conditions. In cancer, hypoxic conditions or molecular alterations within cancer cells can lead to HIF-2α accumulation and promote tumour growth and progression. Inactivating mutations in the von Hippel-Lindau (VHL) gene disable the oxygen-dependent HIF-2α degradation pathway and cause constitutive HIF-2α activity. VHL mutations are prevalent in clear cell renal cell carcinoma (ccRCC) where HIF-2α is a known tumourigenic driver. HIF-2α inhibition was shown to improve ccRCC patient outcomes clinically, warranting development of next-generation inhibitors. EXPERIMENTAL APPROACH Pharmacological effects of a novel small molecule allosteric inhibitor of HIF-2α, AB521 (casdatifan), were evaluated using in vitro cell-based assays and in vivo mouse models. KEY RESULTS AB521 inhibited HIF-2α-mediated transcription in cancer cells, endothelial cells, and M2-polarised macrophages. AB521 was selective for HIF-2α, displaying no activity against HIF-1α, and did not exhibit off-target cytotoxicity. When delivered orally to mice, AB521 caused dose-dependent decreases in HIF-2α-associated pharmacodynamic markers and significant regression of human ccRCC xenograft tumours. AB521 combined favourably with cabozantinib, a standard of care tyrosine kinase inhibitor, or zimberelimab, a clinical-stage anti-PD-1 antibody, in ccRCC xenograft studies. CONCLUSIONS AND IMPLICATIONS AB521 is a potent, selective and orally bioavailable HIF-2α inhibitor, with favourable pharmacological properties, that is being explored clinically for the treatment of ccRCC.
Collapse
Affiliation(s)
| | | | | | | | - Wandi S Zhu
- Arcus Biosciences Inc, Hayward, California, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Román-González A, Califano I, Concepción-Zavaleta M, Pitoia F, Salgado SA. Systemic therapies for medullary thyroid carcinoma: state of the art. Ther Adv Endocrinol Metab 2025; 16:20420188251336091. [PMID: 40356795 PMCID: PMC12066861 DOI: 10.1177/20420188251336091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 04/01/2025] [Indexed: 05/15/2025] Open
Abstract
Medullary thyroid carcinoma (MTC) is a rare neuroendocrine tumor accounting for less than 5% of all thyroid cancers. An estimated 25% of cases are familial secondary to a germline mutation on the rearranged during transfection proto-oncogene (RET); this gene can be present as a somatic mutation in approximately 40%-60% of sporadic MTC tumors. There is an existing genotype-phenotype correlation in the clinical behavior of MTC, with the RET M918T variant associated with aggressive disease. The current systemic treatment profile for progressive metastatic MTC involves antiangiogenics multikinase inhibitors (MKI), specifically cabozantinib and vandetanib, and high-specific RET inhibitor therapy. Decisions on the timing of systemic therapy initiation in this population should involve multidisciplinary care and individualization on a case-by-case scenario; a comprehensive evaluation of performance status, tumor burden, progression rate, medical comorbidities, possible medication interactions, and goals of care must be considered in a patient-centered approach. This review summarizes the evidence on the safety, efficacy, and limitations of systemic therapies for MTC; the aim is to empower clinicians with the knowledge to optimally manage patients with advanced, progressive, or metastatic MTC.
Collapse
Affiliation(s)
- Alejandro Román-González
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, (University of Antioquia), Cra. 51d No. 62-29, Medellín 050001, Colombia
| | - Ines Califano
- Endocrinology Service, Instituto de Oncología “Angel H. Roffo” (Angel H. Roffo Institute of Oncology), University of Buenos Aires, Buenos Aires, Argentina
| | - Marcio Concepción-Zavaleta
- Carrrera de Medicina Humana Universidad Científica del Sur (Scientific University of the South), Lima, Peru
| | - Fabian Pitoia
- Division of Endocrinology, Hospital de Clínicas (Clinics Hospital), University of Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
12
|
Pal SK, Loriot Y, Necchi A, Singh P, Castellano D, Pagliaro L, Suarez C, McGregor BA, Vaishampayan UN, Hauke RJ, Powles T, Van Herpen CM, Courtney KD, Dreicer R, Sudhagoni R, Schwickart M, Andrianova S, Agarwal N. COSMIC-021 Phase Ib Study of Cabozantinib Plus Atezolizumab: Results from the Locally Advanced or Metastatic Urothelial Carcinoma Cohorts. J Clin Oncol 2025; 43:1650-1662. [PMID: 39965176 PMCID: PMC12058374 DOI: 10.1200/jco-24-01675] [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: 08/08/2024] [Revised: 11/13/2024] [Accepted: 12/16/2024] [Indexed: 02/20/2025] Open
Abstract
PURPOSE The COSMIC-021 study assessed the safety and efficacy of cabozantinib plus atezolizumab in advanced solid tumors. Presented here are results from the expansion cohorts with advanced urothelial carcinoma (UC). METHODS This phase Ib study (ClinicalTrials.gov identifier: NCT03170960) enrolled patients with inoperable locally advanced/metastatic UC into four tumor cohorts: first-line cisplatin-eligible (cis-eligible), first-line cisplatin-ineligible (cis-ineligible), previous platinum-containing chemotherapy (previous chemotherapy-treated), and previous immune checkpoint inhibitor (ICI)-treated. Patients received oral cabozantinib 40 mg once daily and intravenous atezolizumab 1,200 mg once every 3 weeks. The primary end point was objective response rate (ORR), as assessed by the investigator per RECIST v1.1 every 6 weeks for 12 months and every 12 weeks thereafter; the secondary end point was safety. RESULTS A total of 121 patients (previous ICI-treated cohort, n = 31, and each of the other cohorts, n = 30) received study treatment from March 2018 to November 2021. The ORR (95% CI) was 30% (15 to 49) for cis-eligible, 20% (8 to 39) for cis-ineligible, 27% (12 to 46) for previous chemotherapy-treated, and 10% (2 to 26) for previous ICI-treated cohorts. The median progression-free survival (95% CI) was 5.5 (1.6 to 11.6), 5.6 (3.1 to 11.1), 5.4 (1.6 to 7.6), and 3.0 (1.8 to 5.5) months, respectively. Grade 3 or 4 treatment-related adverse events (TRAEs) were experienced by 43%, 67%, 57%, and 45% of patients, respectively. TRAEs led to discontinuation of all treatment components in 17%, 13%, 3%, and 19%, respectively. No grade 5 TRAEs were reported in any cohort. CONCLUSION The novel combination of cabozantinib plus atezolizumab exhibited clinical activity in advanced UC that is cis-eligible, cis-ineligible, or previously treated with platinum-containing chemotherapy; clinical activity in previous ICI-treated UC was modest. The toxicity profile was consistent with that previously reported for the combination.
Collapse
Affiliation(s)
| | - Yohann Loriot
- DITEP, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Andrea Necchi
- Department of Medical Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Hospital, Milan, Italy
| | | | | | | | - Cristina Suarez
- Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | | | - Ralph J. Hauke
- Nebraska Cancer Specialists—Midwest Cancer Center, Omaha, NE
| | - Thomas Powles
- Barts Health NHS Trust Saint Bartholomew's Hospital, London, United Kingdom
| | | | | | - Robert Dreicer
- University of Virginia Cancer Center, Charlottesville, VA
| | | | | | | | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
13
|
Tan Z, Völler S, Sancho-Araiz A, Knibbe CAJ, Moes DJAR. A Systematic Evaluation of the Dosing Regimens for Approved Targeted Therapies and Immune Checkpoint Inhibitors in Metastatic Renal Cell Carcinoma From a Project OPTIMUS Perspective. J Clin Pharmacol 2025. [PMID: 40313197 DOI: 10.1002/jcph.70035] [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: 02/10/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025]
Abstract
Targeted therapies and immune checkpoint inhibitors (ICIs) have significantly improved survival outcomes in metastatic renal cell carcinoma (mRCC) but are often associated with high rates of adverse events, leading to dose reductions or treatment discontinuation. The FDA's recent initiative, Project OPTIMUS, emphasizes the importance of optimizing dosing regimens in oncology clinical development, and moves beyond the conventional maximum tolerated dose approach. In this study, we aimed to review and redefine the approved dosing strategies for targeted therapies and ICIs in mRCC from the Project OPTIMUS perspective, including pazopanib, axitinib, cabozantinib, sunitinib, everolimus, and nivolumab. A comprehensive summary of FDA clinical pharmacology reviews and clinical studies performed in routine clinical practice was conducted, alongside model-informed simulations of pharmacokinetic profiles with approved and alternative regimens. Results demonstrated that actual tolerated doses in clinical practice were 46.1% to 86% lower than the approved dosages, with up to 75% of patients requiring dose adjustments. Model-informed simulations suggested that for most targeted therapies, a 14%-50% dose reduction maintained comparable efficacy while improving tolerability. For nivolumab, simulations confirmed adequate drug exposure with the approved flat dose regimens, without an increase of adverse effects. In conclusion, we identified optimized dosing regimens that could improve drug tolerability while maintaining efficacy for approved targeted therapies and ICIs in mRCC. We suggest that these optimized dosing regimens should be considered for use in clinical practice and that the optimal exposure range be included in drug labels to support pharmacokinetically guided dose individualization in clinical practice.
Collapse
Affiliation(s)
- Zhiyuan Tan
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Swantje Völler
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Aymara Sancho-Araiz
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Catherijne A J Knibbe
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Department of Clinical Pharmacy, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
14
|
Gavira J, Auclin E, Rey-Cardenas M, Roy P, Tapia JC, Nay P, Vinceneux A, Lefort F, Nannini S, Randis AMDC, Naoun N, Escudier B, Borchiellini D, de Velasco G, Barthelemy P, Gross-Goupil M, Negrier S, Oudard S, Frazer RD, Albiges L, Flippot R. Activity of lenvatinib-based therapy in previously treated patients with metastatic renal cell carcinoma: A European multicenter study (LENVA-LAT). Eur J Cancer 2025; 220:115389. [PMID: 40184846 DOI: 10.1016/j.ejca.2025.115389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/12/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND AND OBJECTIVE Lenvatinib's activity after immune checkpoint inhibitors (ICI) combination therapy in renal cell carcinoma (RCC) remains unknown. We aimed to describe the real-world outcomes of patients with metastatic RCC (mRCC) treated with lenvatinib after failure of the prior standard of care. METHODS Multicenter retrospective study including patients with mRCC treated with lenvatinib-based therapies beyond first-line therapy between 2020 and 2024. The primary endpoints were objective response rate (ORR) and time-to-treatment failure (TTF). Secondary endpoints included disease control rate (DCR), overall survival (OS), and safety. RESULTS We included 133 patients, with a median age of 61 years. Clear-cell was the main subtype (82.0 %). Before lenvatinib treatment, 15.8 %, 51.9 %, and 27.8 % of patients showed favorable, intermediate, and poor risk disease, respectively, according to the International Metastatic RCC Database Consortium (IMDC). Moreover, patients received a median of 3 previous lines of treatment, including ICIs (90.2 %) and cabozantinib (90.2 %). Lenvatinib was given alone (45.9 %) or in combination with everolimus (33.8 %), pembrolizumab (18.0 %) or investigational agents (2.3 %). The ORR and DCR were 29.1 % and 67.7 %, respectively, with no differences between regimens or lines of treatment. With a median follow-up time of 13.5 months, the median TTF and OS were 6.2 and 9.6 months. Toxicity was manageable with dose modifications required in 34.6 %. The discontinuation rate was 9.8 %, with one toxic death. CONCLUSION Lenvatinib-based regimens were active and safe for heavily pre-treated patients with mRCC. These findings provide evidence to support its use in daily practice.
Collapse
Affiliation(s)
- Javier Gavira
- Department of Medical Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France; Department of Medical Oncology, Institut Català d'Oncologia, Hospitalet de Llobregat, Spain
| | - Edouard Auclin
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Macarena Rey-Cardenas
- Department of Medical Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Pritha Roy
- Department of Clinical Oncology, Velindre Cancer Centre, Velindre University NHS Trust, Cardiff, Wales, United Kingdom
| | - Jose C Tapia
- Department of Medical Oncology, Velindre Cancer Centre, Velindre University NHS Trust, Cardiff, Wales, United Kingdom
| | - Paula Nay
- Department of Medical Oncology, Hôpital Européen Georges-Pompidou, University of Paris, Paris, France
| | - Armelle Vinceneux
- Department of Medical Oncology, Centre Léon Bérard, University of Lyon, Lyon, France
| | - Felix Lefort
- Department of Medical Oncology, Bordeaux University Hospital, Bordeaux, France
| | - Simon Nannini
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | | | - Natacha Naoun
- Department of Medical Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Bernard Escudier
- Department of Medical Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Delphine Borchiellini
- Department of Medical Oncology, Centre Antoine Lacassagne, Université Côte d'Azur, Nice, France
| | - Guillermo de Velasco
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Philippe Barthelemy
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Marine Gross-Goupil
- Department of Medical Oncology, Bordeaux University Hospital, Bordeaux, France
| | - Sylvie Negrier
- Department of Medical Oncology, Centre Léon Bérard, University of Lyon, Lyon, France
| | - Stéphane Oudard
- Department of Medical Oncology, Hôpital Européen Georges-Pompidou, University of Paris, Paris, France
| | - Ricky D Frazer
- Department of Medical Oncology, Velindre Cancer Centre, Velindre University NHS Trust, Cardiff, Wales, United Kingdom
| | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Ronan Flippot
- Department of Medical Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France.
| |
Collapse
|
15
|
Saba NF, Harrington K, Licitra L, Machiels JP, He C, Jew T, Andrianov V, Haddad R. STELLAR-305: phase II/III study of zanzalintinib plus pembrolizumab versus pembrolizumab alone in patients with HNSCC. Future Oncol 2025; 21:1349-1356. [PMID: 40248950 PMCID: PMC12057161 DOI: 10.1080/14796694.2025.2485015] [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: 02/27/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
TWEETABLE ABSTRACT STELLAR-305: design of an ongoing phase III #clinicaltrial, assessing the multi-targeted tyrosine kinase inhibitor, zanzalintinib, in combination with pembrolizumab in previously untreated recurrent/metastatic #HNSCC. CLINICAL TRIAL REGISTRATION NCT06082167 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Nabil F. Saba
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Kevin Harrington
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
- Head and Neck Unit, The Royal Marsden Hospital, London, UK
| | - Lisa Licitra
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Jean-Pascal Machiels
- Institut de Recherche Expérimentale et Clinique, Pôle MIRO, Université catholique de Louvain, Brussels, Belgium
- Department of Medical Oncology, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Brussels, Belgium
| | | | | | | | - Robert Haddad
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
16
|
Jain RK, Swami U, Bilen MA, Gebrael G, Boucher KM, Braun E, Brown JT, Chahoud J, Gupta S, Agarwal N, Sonpavde G, Maughan BL. Cabozantinib plus Pembrolizumab as First-line Therapy for Cisplatin-ineligible Advanced Urothelial Carcinoma: The PemCab Trial. Eur Urol Oncol 2025:S2588-9311(25)00099-9. [PMID: 40307092 DOI: 10.1016/j.euo.2025.04.005] [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: 10/10/2024] [Revised: 03/21/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND AND OBJECTIVE Pembrolizumab monotherapy is approved for patients with platinum-ineligible metastatic urothelial carcinoma (mUC). Cabozantinib is a multireceptor tyrosine kinase inhibitor with activity against MET and VEGFR2 and is approved as monotherapy or in combination with a PD-1 inhibitor for other malignancies. The objective was to determine the safety and efficacy of pembrolizumab + cabozantinib as first-line treatment for patients with mUC. METHODS In this open-label, single-arm, multicenter, phase 2 study, patients received pembrolizumab 200 mg every 3 wk + cabozantinib 40 mg daily. Key inclusion criteria were locally advanced UC or mUC, Eastern Cooperative Oncology Group performance status 0-2, ineligible for or refused cisplatin, and no prior PD-1/L1 inhibitor. The primary endpoint was the objective response rate (ORR). Secondary endpoints included progression-free survival (PFS) and overall survival (OS). According to the statistical plan, in a cohort of 35 evaluable participants, the lower bound of the 95% confidence interval (CI) would extend no more than 26% from the ORR observed, and in a scenario with ≥17 objective responses, the CI would exclude 32%. KEY FINDINGS AND LIMITATIONS Responses were observed in 16 of 35 evaluable patients, with an ORR of 46% (95% CI 31-62%). Median PFS and OS were 8 mo (95% CI 5-13) and 17 mo (95% CI 13-not reached), respectively. The most common treatment-emergent adverse events (any grade) were diarrhea (58%), fatigue (56%), pruritus (39%), nausea (36%), palmar-plantar erythrodysesthesia (36%), and a decrease in appetite (33%). CONCLUSIONS AND CLINICAL IMPLICATIONS This phase 2 trial of pembrolizumab + cabozantinib demonstrated a manageable toxicity profile and promising efficacy as a first-line therapy combination for cisplatin-ineligible patients with mUC.
Collapse
Affiliation(s)
- Rohit K Jain
- Weill Cornell Medicine/NewYork-Presbyterian Hospital, New York, NY, USA
| | - Umang Swami
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Georges Gebrael
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Kenneth M Boucher
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Emma Braun
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | | | - Sumati Gupta
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Guru Sonpavde
- AdventHealth Cancer Institute and University of Central Florida, Orlando, FL, USA
| | | |
Collapse
|
17
|
Assi A, Farhat M, Mohanna R, Hachem MCR, Zalaquett Z, Aoun M, Farraj SA, Daher M, Sebaaly A, Kourie HR. Tyrosine kinase inhibitors in Ewing's sarcoma: a systematic review. BMC Cancer 2025; 25:735. [PMID: 40251562 PMCID: PMC12008964 DOI: 10.1186/s12885-025-14130-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
Ewing's sarcoma (ES) is a highly aggressive primary bone malignancy that primarily affects children and adolescents. Several tyrosine kinase receptors (RTKs) have been found to be overexpressed in ES samples, and it was demonstrated that some play significant roles in driving the malignant phenotype of ES. Specifically, ES with insulin-like growth factor 1 (IGF1R) or vascular endothelial growth factor (VEGFR) overexpression were correlated with more aggressive ES and worse outcomes. Other RTKs that were determined to be overexpressed in ES include platelet-derived growth factor receptor, stem cell factor receptor, and hepatocyte growth factor. Overexpression of these molecules suggests their possible tumor-driving role, making them potential targets for intervention. Various tyrosine kinase inhibitors (TKIs), including apatinib, anlotinib, and cabozantinib have shown clinical promise in patients with recurrent ES who have progressed on previous lines of therapy. The findings reported in this review emphasize the importance of assessing IGF1R-focused inhibitors and combinational therapeutic regimens in future research. Furthermore, biomarkers predictive of response are necessary to improve patient outcomes. In order to optimize ES care, considerations for patient eligibility on the basis of positivity for biomarkers predictive of response, and the inclusion of quality-of-life evaluations in studies must be addressed.
Collapse
Affiliation(s)
- Ahmad Assi
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon.
| | - Mohamad Farhat
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon
| | - Rami Mohanna
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon
| | | | - Ziad Zalaquett
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon
| | - Marven Aoun
- Orthopedics Department, Hotel Dieu de France, Beirut, Lebanon
| | - Sami Abi Farraj
- Hematology-Oncology Department, Hotel Dieu de France, Beirut, Lebanon
| | - Mohammad Daher
- Orthopedics Department, Hotel Dieu de France, Beirut, Lebanon.
- Orthopedics Department, Brown University, Providence, RI, USA.
| | - Amer Sebaaly
- Orthopedics Department, Hotel Dieu de France, Beirut, Lebanon.
| | | |
Collapse
|
18
|
Gil-Bernabé S, García-DeLaFuente L, García-Rostán G. The Revolution of Targeted Therapies in Thyroid Cancer Treatment: Present and Future Promising Anti-Cancer Drugs. Int J Mol Sci 2025; 26:3663. [PMID: 40332222 PMCID: PMC12027515 DOI: 10.3390/ijms26083663] [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: 02/19/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 05/08/2025] Open
Abstract
Thyroid cancer prevalence has increased in the last few decades. Whereas the majority of well-differentiated histotypes have effective therapeutic options, the most advanced cases lacked successful treatment until recent years. Genomic alterations have emerged as targets for new anti-cancer drugs. This molecular knowledge is gradually being translated into sophisticated approaches for the stratification, management, and therapies of patients with thyroid carcinomas. The genomic characterisation of tumours in clinical assistance serves as a tool for enhancing the prognostic assessment of patients with thyroid cancer and predicting their responses to the agents. The MAPK pathway is the most predominantly activated molecular route in this cancer. Several drugs have been developed to inhibit this pathway at different levels. However, the acquired resistance that emerges is the main problem in their use. Other strategies targeting not only driver mutations but also those that confer aggressive behaviour on tumours can be potential targetable options. Due to the new therapies, patients with the most aggressive histotypes have improved survival rates. Adverse events, although manageable, have a high prevalence among the current therapies. Selective inhibitors, immunotherapies, and the combination of both will play a pivotal role in the treatment and the improvements in overall survival in thyroid cancer patients.
Collapse
Affiliation(s)
- Sara Gil-Bernabé
- Pathology Department, Faculty of Medicine, Valladolid University, 47003 Valladolid, Spain
- Group Pathobiology of Cancer: Inter-, Intra-Tumor Heterogeneity and Molecular Targets, Institute of Molecular Genetics and Biomedicine (IBGM), 47003 Valladolid, Spain
| | | | - Ginesa García-Rostán
- Pathology Department, Faculty of Medicine, Valladolid University, 47003 Valladolid, Spain
- Group Pathobiology of Cancer: Inter-, Intra-Tumor Heterogeneity and Molecular Targets, Institute of Molecular Genetics and Biomedicine (IBGM), 47003 Valladolid, Spain
| |
Collapse
|
19
|
Tsai YC, Li JR, Chiu KY, Su PJ, Su YL, Chung HJ, Li CC, Huang CP, Guo JC, Chen CS, Chang I, Perrot V, Chang YH. Real-world study of cabozantinib treatment of advanced renal cell carcinoma in Taiwan. J Formos Med Assoc 2025:S0929-6646(25)00117-2. [PMID: 40221295 DOI: 10.1016/j.jfma.2025.03.016] [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: 11/28/2023] [Revised: 09/27/2024] [Accepted: 03/14/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND There is a lack of real-world evidence from Taiwan on the use of cabozantinib for advanced renal cell carcinoma (aRCC). We evaluated cabozantinib treatment for aRCC after previous antiangiogenic therapy in real-world Taiwanese clinical practice. METHODS Medical records from seven Taiwanese hospitals were retrospectively analyzed. Eligible patients were adults with aRCC who initiated cabozantinib between October 2018 and August 2021 after ≥1 antiangiogenic therapy. Patient characteristics and treatment patterns were described. Outcomes included objective response rate (ORR [complete or partial]; primary endpoint) assessed by RECIST v1.1 or local investigator assessment, progression-free survival (PFS), and tolerability. RESULTS Fifty-one patients were included: 39.2 % received cabozantinib second-line, 33.3 % third-line, and 27.5 % fourth- or later-line. Mean patient age was 61.2 years; most were male (80 %), had clear-cell (80 %), metastatic (92 %) disease, and had previous nephrectomy (78 %). Sunitinib and pazopanib were the most common previous (any line) treatments (63 % and 53 % of patients, respectively). Dose reductions occurred in 47 % of patients and were more common in patients (57 %) who initiated cabozantinib at 60 mg/day than in those (33 %) who initiated at 40 mg/day (72 % vs 12 %, respectively); discontinuation rates were similar for these groups (48 % vs 47 %, respectively). Overall, ORR was 39.2 % (95 % CI: 25.8, 53.9) and median PFS was 12.4 months (95 % CI: 8.2, 16.3). Rates of serious treatment-emergent adverse events related to cabozantinib were low (7.8 %). CONCLUSION This Taiwanese study found the real-world effectiveness and tolerability of cabozantinib after previous antiangiogenic therapy to be consistent with the results of the METEOR trial.
Collapse
Affiliation(s)
- Yu-Chieh Tsai
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jian-Ri Li
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Kun-Yuan Chiu
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Po-Jung Su
- Division of Hematology-Oncology, Chang Gung Memorial Hospital Linkou, Linkou, and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Li Su
- Division of Hematology Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Cancer Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Genomic & Proteomic Core Lab, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsiao-Jen Chung
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Urology, College of Medicine and Shu-Tien Urological Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ching-Chia Li
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chi-Ping Huang
- Department of Urology, China Medical University and Hospital, Taichung, Taiwan
| | - Jhe-Cyuan Guo
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chuan-Shu Chen
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | | | | | - Yen-Hwa Chang
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
20
|
Fujiwara Y, Kuroda H, Abe T, Endo K, Oikawa T, Tsuruoka M, Ninomiya M, Fujita M, Abe K, Katsumi T, Minami S, Sato W, Igarashi G, Iino C, Tanabe N, Numao H, Kimura O, Nakaya I, Ito A, Watanabe T, Yusa K, Nagasawa T, Sato H, Suzuki A, Yoshida Y, Sawara K, Kakisaka K, Miyasaka A, Ohira H, Ueno Y, Matsumoto T. Optimizing Cabozantinib Dosing in Unresectable Hepatocellular Carcinoma of 7-on/7-off Regimen. Cancers (Basel) 2025; 17:1288. [PMID: 40282464 PMCID: PMC12025779 DOI: 10.3390/cancers17081288] [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: 03/20/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Cabozantinib (CAB) causes a high incidence of adverse events in unresectable hepatocellular carcinoma (u-HCC) and requires dose adjustments. We evaluated the efficacy and safety of the 7-on/7-off regimen composed of 7 consecutive days' administration of CAB followed by a 7-day rest period. This was a retrospective analysis of 35 patients with u-HCC, who were treated with CAB in a multicenter cohort in Japan from 2020 to 2024. The clinical outcomes of 12 patients treated with the 7-on/7-off regimen and 23 patients treated with daily dosing were compared. There were significant differences in overall survival (12.4 months vs. 6.3 months, p = 0.03), median progression-free survival (4.8 months vs. 3.2 months, p < 0.01), objective response rate (16.7% vs. 0.0%, p = 0.04), and incidence of any adverse events (75.0% vs. 100.0%, p = 0.03) between the 7-on/7-off regimen group and daily dosing group. The median duration of drug exposure (122 days vs. 42 days, p < 0.01) and median duration of dose reduction (100 days vs. 23 days, p < 0.01) were prolonged significantly in the 7-on/7-off group than in the daily dosing group. CAB in a 7-on/7-off regimen may improve the tolerability and treatment response in patients with u-HCC.
Collapse
Affiliation(s)
- Yudai Fujiwara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Hidekatsu Kuroda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Tamami Abe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Kei Endo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Takayoshi Oikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Mio Tsuruoka
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi 9808574, Japan
| | - Masashi Ninomiya
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi 9808574, Japan
| | - Masashi Fujita
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 96012950, Japan
| | - Kazumichi Abe
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 96012950, Japan
| | - Tomohiro Katsumi
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, Yamagata 9902311, Japan
| | - Shinichiro Minami
- Department of Gastroenterology, Graduate School of Medicine, Akita University, Akita 0100041, Japan
| | - Wataru Sato
- Department of Gastroenterology, Graduate School of Medicine, Akita University, Akita 0100041, Japan
| | - Go Igarashi
- Department of Gastroenterology, Hirosaki University Graduate School of Medicine, Aomori 0368563, Japan
| | - Chikara Iino
- Department of Gastroenterology, Hirosaki University Graduate School of Medicine, Aomori 0368563, Japan
| | - Nobukazu Tanabe
- Department of Gastroenterology, National Hospital Organization Sendai Medical Center, Miyagi 9830045, Japan
| | - Hiroshi Numao
- Department of Gastroenterology, Aomori Prefectural Central Hospital, Aomori 0300913, Japan
| | - Osamu Kimura
- Department of Gastroenterology, South Miyagi Medical Center, Miyagi 9891253, Japan
| | - Ippeki Nakaya
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Asami Ito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Takuya Watanabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Kenji Yusa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Tomoaki Nagasawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Hiroki Sato
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Akiko Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Yuichi Yoshida
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Kei Sawara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Keisuke Kakisaka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Akio Miyasaka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| | - Hiromasa Ohira
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 96012950, Japan
| | - Yoshiyuki Ueno
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, Yamagata 9902311, Japan
| | - Takayuki Matsumoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate 0283695, Japan
| |
Collapse
|
21
|
Elkotamy MS, Elgohary MK, Elkelesh IA, Alkabbani MA, Khaleel EF, Eldehna WM, Abdel-Aziz HA. Design, synthesis, and molecular dynamics-driven evaluation of quinoline-sulfonamide derivatives as potent and selective EGFR inhibitors with promising anti-cancer efficacy and safety profiles. Bioorg Chem 2025; 157:108247. [PMID: 39983403 DOI: 10.1016/j.bioorg.2025.108247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 02/23/2025]
Abstract
The creation of new molecules that target EGFR is essential for the progression of cancer treatment. This study synthesized and evaluated 16 quinoline-sulfonamide derivatives for their potential as anti-cancer agents. Compound 8c, which contains a methoxy group on the benzenesulfonamide tail, exhibited notable EGFR inhibitory activity (IC50 = 0.161 µM), similar to that of Erlotinib (IC50 = 0.142 µM). Compound 8c demonstrated enhanced in-vitro cytotoxicity against HCT-116, MCF-7, HeLa, and HepG2 cancer cell lines. Studies on the cell cycle and apoptosis demonstrated that compound 8c caused G1/S arrest and markedly enhanced apoptosis in HepG2 cells. In-vivo, compound 8c demonstrated comparable and/or superior efficacy compared to doxorubicin in decreasing tumor volume, weight, TNF-alpha, and COX-2 levels in the SEC model, alongside improved histopathological and immunohistochemical results. Molecular docking and dynamic simulations confirmed its stable binding to EGFR, exhibiting superior stability metrics in comparison to Erlotinib. Pharmacokinetic and toxicity evaluations indicated that compound 8c exhibits favorable drug-like properties and a safer toxicity profile. These findings identify compound 8c as a potential candidate for the development of safe and effective anti-cancer therapies, necessitating additional preclinical investigations.
Collapse
Affiliation(s)
- Mahmoud S Elkotamy
- Pharmaceutical Chemistry Department Faculty of Pharmacy Egyptian-Russian University Badr City 11829 Cairo Egypt.
| | - Mohamed K Elgohary
- Pharmaceutical Chemistry Department Faculty of Pharmacy Egyptian-Russian University Badr City 11829 Cairo Egypt
| | - Islam A Elkelesh
- Pharmaceutical Chemistry Department Faculty of Pharmacy Egyptian-Russian University Badr City 11829 Cairo Egypt
| | - Mahmoud Abdelrahman Alkabbani
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo 11829 Egypt
| | - Eman F Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Asir 61421 Saudi Arabia
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh P.O. Box 33516, Egypt.
| | - Hatem A Abdel-Aziz
- Applied Organic Chemistry Department, National Research Center, Dokki 12622, Cairo, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria 21648 Egypt.
| |
Collapse
|
22
|
Chiu CL, Zhang D, Zhao H, Wei Y, Polasko AL, Thomsen MT, Yang V, Yang KK, Hauck S, Peterson EE, Wen RM, Qiu Z, Corey E, Miao YR, Rankin EB, Peehl DM, Huang J, Giaccia AJ, Brooks JD. Targeting AXL Inhibits the Growth and Metastasis of Prostate Cancer in Bone. Clin Cancer Res 2025; 31:1346-1358. [PMID: 39879384 PMCID: PMC11961319 DOI: 10.1158/1078-0432.ccr-24-3028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/22/2024] [Accepted: 01/27/2025] [Indexed: 01/31/2025]
Abstract
PURPOSE After failing primary and secondary hormonal therapy, castration-resistant and neuroendocrine prostate cancer metastatic to the bone is invariably lethal, although treatment with docetaxel and carboplatin can modestly improve survival. Therefore, agents targeting biologically relevant pathways in prostate cancer and potentially synergizing with docetaxel and carboplatin in inhibiting bone metastasis growth are urgently needed. EXPERIMENTAL DESIGN Phosphorylated (activated) AXL expression in human prostate cancer bone metastases was assessed by IHC staining. We evaluated the effects of a novel soluble AXL signaling inhibitor, sAXL (batiraxcept or AVB-S6-500), on tumor growth and lung metastases in prostate cancer patient-derived xenograft models that were implanted intratibially. After injection of LuCaP cells into the tibiae, tumors were treated with batiraxcept and docetaxel or carboplatin alone or in combination, and tumor growth was monitored by serum prostate-specific antigen or bioluminescence. Tumor burden was quantified by human-specific Ku70 staining, and metastasis to the lungs was determined using qPCR. Transcriptomic profiling, Western blotting, and immunohistochemistry were performed to identify treatment-regulated gene and protein profile changes. RESULTS High AXL phosphorylation in human prostate cancer bone metastases correlated with shortened survival. Batiraxcept alone or in combination with docetaxel or carboplatin significantly suppressed intratibial tumor growth and suppressed metastasis to the lungs through multiple mechanisms, including repression of cancer stemness genes (CD44, ALDH1A1, TACSTD2, and ATXN1) and the PI3K, JAK, MAPK, and E2F1/NUSAP1 signaling pathways. CONCLUSIONS Our study provides a robust preclinical rationale and mechanisms of action for using batiraxcept as a single agent or in combination with docetaxel or carboplatin to treat lethal metastatic prostate cancer.
Collapse
Affiliation(s)
- Chun-Lung Chiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Dalin Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yi Wei
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Mikkel Thy Thomsen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Clinical Medicine, Aarhus University, Denmark
| | - Vanessa Yang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kasie Kexin Yang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Spencer Hauck
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Eric E. Peterson
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ru M. Wen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhengyuan Qiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Yu Rebecca Miao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erinn B. Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Donna M. Peehl
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Amato J. Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Oncology, University of Oxford, Oxford, UK
| | - James D. Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
23
|
Elgammal WE, Elkady H, Dahab MA, Mahdy HA, Hagras M, Nofal A, Alsfouk BA, Elkaeed EB, Eissa IH, Metwaly AM. Design and synthesis of thiadiazoles as anticancer, apoptotic, and VEGFR-2 inhibitors. Future Med Chem 2025; 17:915-927. [PMID: 40197130 DOI: 10.1080/17568919.2025.2485863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Vascular endothelial growth factor receptor (VEGFR-2) inhibitors are critical in cancer therapy due to their role in suppressing tumor angiogenesis. Herein, we report a new series of thiadiazole-based derivatives as potential VEGFR-2 inhibitors with promising anticancer activity. METHODS The synthesized compounds were evaluated for anti-proliferative activity against human cancer cell lines (HCT-116, MCF-7, and HepG-2), and WI-38 as normal cells. Sorafenib was used as a reference drug. VEGFR-2 inhibitory activity was determined, followed by cell cycle analysis, apoptosis assays, Q-RT-PCR analysis, and wound-healing assays. In silico molecular docking was conducted to explore binding interactions. RESULTS Among the tested compounds, 13b exhibited potent anti-proliferative activity (IC50: 3.98-11.81 µM) and strong VEGFR-2 inhibition (IC50: 41.51 nM), surpassing sorafenib (IC50: 53.32 nM). Cell cycle analysis revealed that 13b induced G2/M phase arrest in MCF-7 cells. Apoptosis levels increased from 2% to 52%, accompanied by a > 12-fold rise in the Bax/Bcl-2 ratio and activation of caspase-8/9. Additionally, 13b significantly suppressed MCF-7 cell migration, with only 5.28% wound closure. In silico studies confirmed its strong VEGFR-2 binding interactions. CONCLUSION Thiadiazole-based derivatives, particularly compound 13b, exhibit potent VEGFR-2 inhibition, anti-proliferative effects, apoptosis induction, and anti-migratory activity, supporting their potential as promising anticancer agents.
Collapse
Affiliation(s)
- Walid E Elgammal
- Department of Chemistry, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Hazem Elkady
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Mohammed A Dahab
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Hazem A Mahdy
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Mohamed Hagras
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Ahmed Nofal
- Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Bshra A Alsfouk
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Eslam B Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ahmed M Metwaly
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
24
|
Wang Z, Zheng F, Wan L, Zhang L, Xiong S, Li S, Wang C, Liu X, Deng J. Safety assessment of cabozantinib in patients with renal cell carcinoma: retrospective pharmacovigilance study based on FAERS database. Expert Opin Drug Saf 2025; 24:427-433. [PMID: 39545449 DOI: 10.1080/14740338.2024.2429475] [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: 09/14/2024] [Revised: 10/31/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE This study was designed to conduct data mining through the Food and Drug Administration Adverse Event Reporting System (FAERS) to assess adverse events (AEs) associated with cabozantinib in the treatment of renal cell carcinoma. METHODS Reporting odds ratio (ROR), proportional reporting ratio (PRR), Bayesian confidence propagation neural network (BCPNN), and multi-item gamma Poisson shrinker (MGPS) algorithms were used to detect drug-related AEs signals from reporting data in FAERS database from 2016 to 2024. RESULTS A total of 32,129 AE reports identifying cabozantinib as a 'primary suspect' were retrieved from the FAERS database. Among them, there were 21,549 reports of renal cell carcinoma as an indication. AEs induced by cabozantinib were observed in 23 system organ classes (SOCs). 215 AE signals were detected in 16 SOCs after four algorithms were simultaneously met. Among them, signals related to gastrointestinal disorders, general disorders and administration site conditions, and skin and subcutaneous tissue disorders were the most common. Of note, the median time to onset of AEs was 38 days (interquartile range (IQR) 14-116 days). CONCLUSION This study provides new insights into the monitoring, surveillance, and management of cabozantinib-related adverse drug reactions and provides a comprehensive long-term post-marketing safety assessment of cabozantinib.
Collapse
Affiliation(s)
- Zhipeng Wang
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Jiangxi Institute of Urology, Nanchang, China
| | - Fuchun Zheng
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Jiangxi Institute of Urology, Nanchang, China
| | - Liangwei Wan
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Jiangxi Institute of Urology, Nanchang, China
| | - Lei Zhang
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Jiangxi Institute of Urology, Nanchang, China
| | - Situ Xiong
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Jiangxi Institute of Urology, Nanchang, China
| | - Sheng Li
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Jiangxi Institute of Urology, Nanchang, China
| | - Chen Wang
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Jiangxi Institute of Urology, Nanchang, China
| | - Xiaoqiang Liu
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Jiangxi Institute of Urology, Nanchang, China
| | - Jun Deng
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Jiangxi Institute of Urology, Nanchang, China
| |
Collapse
|
25
|
Sattler M, Salgia R. The expanding role of the receptor tyrosine kinase MET as a therapeutic target in non-small cell lung cancer. Cell Rep Med 2025; 6:101983. [PMID: 40020676 PMCID: PMC11970332 DOI: 10.1016/j.xcrm.2025.101983] [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] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 03/03/2025]
Abstract
Aberrant regulation of MET receptor tyrosine kinase activity is a frequent event in non-small cell lung cancer (NSCLC), even though the frequency of oncogenic driver mutations of MET is low. Our discovery of oncogenic MET exon 14 skipping mutations, the characterization of the first prototype MET kinase inhibitor, and characterization of MET expression levels have led the way to novel therapeutic approaches with improved outcomes in NSCLC. MET exon 14 mutations are the most consequential but not the only alterations that can be targeted through small molecule tyrosine kinase inhibitors. The abundant expression of cellular MET (c-MET) in cancer cells has provided new opportunities for immuno-oncology approaches in a broader patient population, and the integration of MET-targeted personalized medicine with immunotherapy has not been fully exploited yet. Here, we highlight essential facets of MET as a therapeutic target in NSCLC and provide an outlook for future approaches.
Collapse
Affiliation(s)
- Martin Sattler
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
26
|
de Andrade FA, Bulzico D, Corbo R, Vaisman F. Is peptide receptor radionuclide therapy still a promising option for medullary thyroid carcinoma? Endocrine 2025; 87:943-950. [PMID: 39609369 DOI: 10.1007/s12020-024-04114-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/13/2024] [Indexed: 11/30/2024]
Abstract
Medullary thyroid carcinoma (MTC) is a rare cancer that originates from germline RET proto-oncogene mutations in all hereditary forms and from somatic RET mutations in most sporadic cases. Currently, highly selective RET inhibitors have been approved for clinical use in patients with RET mutations with persistent, recurrent or metastatic disease. This therapy has proven efficacy, low toxicity, and a limited impact on patients' quality of life. However, for recurrent or metastatic RET-negative disease, few systemic therapies are available. Multikinase inhibitors are used; however, tumour cells frequently develop resistance mechanisms, or treatment must be discontinued due to the high incidence of side effects. In this context, peptide receptor radionuclide therapy (PRRT) may be a treatment option, but its clinical utility remains under investigation. The aim of this review is to evaluate the evidence of PRRT in MTC and discuss its limitations in the RET inhibitor era.
Collapse
Affiliation(s)
| | - Daniel Bulzico
- Endocrine Oncology Unit, Brazilian National Cancer Institute, INCA, Rio de Janeiro, Brazil
- Nuclear Medicine Section, Brazilian National Cancer Institute, INCA, Rio de Janeiro, Brazil
| | - Rossana Corbo
- Endocrine Oncology Unit, Brazilian National Cancer Institute, INCA, Rio de Janeiro, Brazil
- Nuclear Medicine Section, Brazilian National Cancer Institute, INCA, Rio de Janeiro, Brazil
| | - Fernanda Vaisman
- Endocrine Oncology Unit, Brazilian National Cancer Institute, INCA, Rio de Janeiro, Brazil.
- Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil.
| |
Collapse
|
27
|
Bilen MA, Vo BT, Liu Y, Greenwald R, Davarpanah AH, McGuire D, Shiradkar R, Li L, Midya A, Nazha B, Brown JT, Williams S, Session W, Russler G, Caulfield S, Joshi SS, Narayan VM, Filson CP, Ogan K, Kucuk O, Carthon BC, Del Balzo L, Cohen A, Boyanton A, Prokhnevska N, Cardenas MA, Sobierajska E, Jansen CS, Patil DH, Nicaise E, Osunkoya AO, Kissick HT, Master VA. Neoadjuvant cabozantinib for locally advanced nonmetastatic clear cell renal cell carcinoma: a phase 2 trial. NATURE CANCER 2025; 6:432-444. [PMID: 40016487 DOI: 10.1038/s43018-025-00922-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 01/29/2025] [Indexed: 03/01/2025]
Abstract
Cabozantinib is an oral multikinase inhibitor approved for treatment in metastatic renal cell carcinoma (RCC). We conducted a phase 2, nonrandomized, single-arm clinical trial (NCT04022343) of cabozantinib treatment for 12 weeks in 17 patients with locally advanced, biopsy-proven, nonmetastatic clear cell RCC before surgical resection. The primary end point was the objective response rate (complete and partial responses) at week 12 and secondary end points included safety, tolerability, clinical and surgical outcomes, and quality of life. Six patients (35%) experienced a partial response and 11 patients (65%) had stable disease. The most common adverse events were diarrhea (n = 12, 70.6%), anorexia, fatigue and hypertension (n = 10, 58.8%), nausea and palmar-plantar erythrodysesthesia syndrome (n = 9, 52.9%). No treatment grade 4 or 5 adverse events related to cabozantinib or surgery occurred. The 1-year disease-free survival and overall survival were 82.4% (95% CI 54.7-93.9%) and 94.1% (95% CI 65-99.1%), respectively. Cabozantinib treatment activated CD8+ T cells in the blood, depleted myeloid populations and induced immune niches for TCF1+ stem-like CD8+ T cells. Cabozantinib was clinically active and safe in the neoadjuvant setting in patients with locally advanced nonmetastatic clear cell RCC.
Collapse
Affiliation(s)
- Mehmet A Bilen
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.
| | - BaoHan T Vo
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuan Liu
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Rachel Greenwald
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Amir H Davarpanah
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Donald McGuire
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Rakesh Shiradkar
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Liping Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Adhishek Midya
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Bassel Nazha
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jacqueline T Brown
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sierra Williams
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Wilena Session
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Greta Russler
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Sarah Caulfield
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Pharmaceutical Services, Emory University School of Medicine, Atlanta, GA, USA
| | - Shreyas S Joshi
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Vikram M Narayan
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Kenneth Ogan
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Omer Kucuk
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Bradley Curtis Carthon
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Luke Del Balzo
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Athena Cohen
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Adriana Boyanton
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Ewelina Sobierajska
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Caroline S Jansen
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dattatraya H Patil
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Edouard Nicaise
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Adeboye O Osunkoya
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Haydn T Kissick
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Vaccine Center, Emory University, Atlanta, GA, USA.
| | - Viraj A Master
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
28
|
Buzatu IM, Tataranu LG, Duta C, Stoian I, Alexandru O, Dricu A. A Review of FDA-Approved Multi-Target Angiogenesis Drugs for Brain Tumor Therapy. Int J Mol Sci 2025; 26:2192. [PMID: 40076810 PMCID: PMC11899917 DOI: 10.3390/ijms26052192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Neovascularization is an important process in brain tumor development, invasion and metastasis. Several research studies have indicated that the VEGF signaling target has potential for reducing angiogenesis in brain tumors. However, targeting VEGF signaling has not met the expected efficacy, despite initial enthusiasm. This is partly because tumors cleverly use alternative growth factor pathways, other than VEGF signaling, to restore angiogenesis. Multi-target inhibitors have been developed to inhibit several receptor kinases that play a role in the development of angiogenesis. By simultaneously affecting various receptor kinases, these treatments can potentially obstruct various angiogenic pathways that are involved in brain cancer advancement, often offering a more holistic strategy than treatments focusing on just one kinase. Since 2009, the FDA has approved a number of multi-kinase inhibitors that target angiogenic growth factor receptors (e.g., VEGFR, PDGFR, FGFR, RET, c-KIT, MET, AXL and others) for treatment of malignant diseases, including brain cancer. Here, we present some recent results from the literature regarding the preclinical and clinical effects of these inhibitors on brain tumors.
Collapse
Affiliation(s)
- Iuliana Mihaela Buzatu
- Department of Microbiology, “Fundeni” Clinical Institute, Șoseaua Fundeni 258, 022328 Bucharest, Romania;
| | - Ligia Gabriela Tataranu
- Department of Neurosurgery, Clinical Emergency Hospital “Bagdasar-Arseni”, Soseaua Berceni 12, 041915 Bucharest, Romania;
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Carmen Duta
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania; (C.D.); (I.S.); (A.D.)
| | - Irina Stoian
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania; (C.D.); (I.S.); (A.D.)
| | - Oana Alexandru
- Department of Neurology, University of Medicine and Pharmacy of Craiova, Petru Rares 2, 200349 Craiova, Romania
| | - Anica Dricu
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania; (C.D.); (I.S.); (A.D.)
| |
Collapse
|
29
|
Liu Z, Liu W, Shen X, Jiang T, Li X, Liu H, Zheng Z. Molecular mechanism of type ib MET inhibitors and their potential for CNS tumors. Sci Rep 2025; 15:6926. [PMID: 40011494 PMCID: PMC11865562 DOI: 10.1038/s41598-025-85631-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/06/2025] [Indexed: 02/28/2025] Open
Abstract
The emergence of targeted therapies for MET exon 14 (METex14) skipping mutations has significantly changed the treatment landscape for NSCLC and other solid tumors. The skipping of METex14 results in activating the MET-HGF pathway and promoting tumor cell proliferation, migration, and preventing apoptosis. Type Ib MET inhibitors, designed to selectively target the "DFG-in" conformation of MET, characteristically bind to the ATP-binding pocket of MET in a U-shaped conformation, extending into the solvent-accessible region and interact strongly with residue Y1230 through π-π interactions, have shown remarkable efficacy in treating METex14-altered NSCLC, including cases with brain metastases (BMs). Notably, vebreltinib and capmatinib have demonstrated superior blood-brain barrier (BBB) permeability in both computational and experimental models, highlighting their potential for treating the central nervous system (CNS) metastases. P-glycoprotein (P-gp) is highly expressed in the BBB, which limits the brain uptake of many highly lipophilic drugs. Despite challenges posed by P-gp mediated efflux, vebreltinib has emerged as a promising candidate for CNS treatment due to its favorable pharmacokinetic profile and minimal susceptibility to P-gp efflux. This study underscores the importance of molecular dynamics simulations in predicting drug efficacy and BBB penetration, providing valuable insights for the development of CNS-targeted metastases therapies.
Collapse
Affiliation(s)
- Zhenhao Liu
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Wenlang Liu
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Xinyi Shen
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Tao Jiang
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Xionghao Li
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Hao Liu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, Hubei, People's Republic of China.
| | - Zheng Zheng
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
30
|
Khan E, Hylton H, Rajan N, Bouley SJ, Siddiqui JK, Rajasekaran S, Koshre GR, Storts H, Valenciaga A, Khan M, Liyanarachchi S, Fernandez F, Zheng X, Phay J, Dedhia PH, Wang J, Walker JA, Ringel MD, Miles WO. Proteomic Profiling of Medullary Thyroid Cancer Identifies CAPN1 as a Key Regulator of NF1 and RET Fueled Growth. Thyroid 2025; 35:177-187. [PMID: 39868924 DOI: 10.1089/thy.2024.0102] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Background: Medullary thyroid cancer (MTC) is a frequently metastatic tumor of the thyroid that develops from the malignant transformation of C-cells. These tumors most commonly have activating mutations within the RET or RAS proto-oncogenes. Germline mutations within RET result in C-cell hyperplasia, and cause the MTC pre-disposition disorder, multiple endocrine neoplasia, type 2A (MEN2A). Single-agent therapies for MTC, including vandetanib (VAN) and cabozantinib for all MTCs and selpercatinib (SEL) for RET-mutated MTC, lead to partial responses but are not curative. Methods: To identify new therapeutic targets for MTC, we conducted proteomic profiling of normal C-cells, MTC cells, pre-malignant MEN2A patient samples, and MTC tumors. Results: From this analysis, we identified CAPN1, a member of the CALPAIN (CAPN) family endopeptidases, as widely upregulated in MTC samples. We found that short hairpin RNA-mediated depletion of CAPN1 or inhibitors of CAPN1 significantly reduced MTC cell growth, colony formation, and xenograft tumor growth in vivo. In addition, we show that CAPN1 inhibitors synergize with VAN and SEL in vitro, maximizing apoptosis. Mechanistic experiments implicate CAPN1 in inhibiting neurofibromin, encoded by NF1, and CAPN1 inhibitors stabilize NF1 protein levels and diminish downstream RAS/RET activation of AKT and ERK. Conclusions: Our data suggest that increased CAPN1 levels support RET and RAS-fueled growth by reducing NF1 levels. We find that combinatorial therapies between CAPN1 inhibitors and VAN or SEL show maximal efficacy in MTC cells.
Collapse
Affiliation(s)
- Eshan Khan
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Hannah Hylton
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Neel Rajan
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Department of Molecular Medicine and Therapeutics, The Ohio State University, Columbus, Ohio, USA
| | - Stephanie J Bouley
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jalal K Siddiqui
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Swetha Rajasekaran
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Ganesh R Koshre
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Hayden Storts
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Anisley Valenciaga
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Department of Molecular Medicine and Therapeutics, The Ohio State University, Columbus, Ohio, USA
| | - Misbah Khan
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Sandya Liyanarachchi
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Department of Molecular Medicine and Therapeutics, The Ohio State University, Columbus, Ohio, USA
| | - Francisco Fernandez
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xuguang Zheng
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - John Phay
- Division of Surgical Oncology, Ohio State University Comprehensive Cancer Center and Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Priya H Dedhia
- Division of Surgical Oncology, Ohio State University Comprehensive Cancer Center and Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Jing Wang
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - James A Walker
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Matthew D Ringel
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Department of Molecular Medicine and Therapeutics, The Ohio State University, Columbus, Ohio, USA
| | - Wayne O Miles
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
31
|
Santoni M, Mollica V, Rizzo A, Massari F. Dynamics of resistance to immunotherapy and TKI in patients with advanced renal cell carcinoma. Cancer Treat Rev 2025; 133:102881. [PMID: 39799795 DOI: 10.1016/j.ctrv.2025.102881] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
Immune-based combinations are the cornerstone of the first-line treatment of metastatic renal cell carcinoma patients, leading to outstanding outcomes. Nevertheless, primary resistance and disease progression is a critical clinical challenge. To properly address this issue, it is pivotal to understand the mechanisms of resistance to immunotherapy and tyrosine kinase inhibitors, that tumor eventually develop under treatment. In this review of the literature, we aim at exploring resistance mechanisms arising in patients treated with first-line immune-based combinations in order to understand the biological pattern that should be investigated to overcome them. In more detail, mechanisms of resistance to nivolumab and pembrolizumab are divided into intrinsic to cancer cells and extrinsic (stromal or immune cells). Regarding axitinib, the increased expression of Nuclear protein 1 (NUPR1) or decreased levels of insulin receptor (INSR) characterize resistant cells. The secretion of non-VEGF pro-angiogenic factors, such as PDGF-BB, IL-1β, MMP-9, Gro-α, IL-8, IL-6, and CCL-2, can lead to resistance to cabozantinib. The reactivation of pathways previously targeted by lenvatinib or the activation of alternative pathways, such as EGFR-PAK2-ERK pathway, underlie the development of resistance to lenvatinib. Exploring resistance mechanism that arise during first-line therapy can lead to the development of treatment strategy able to overcome them in order to improve duration of response and patients outcomes.
Collapse
Affiliation(s)
- Matteo Santoni
- Medical Oncology Unit, Macerata Hospital, Macerata, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy.
| |
Collapse
|
32
|
Melhorn P, Raderer M, Kiesewetter B. Selecting systemic treatment for metastatic neuroendocrine tumors of the lung-current evidence and clinical implications. Cancer Treat Rev 2025; 133:102878. [PMID: 39787793 DOI: 10.1016/j.ctrv.2024.102878] [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: 09/20/2024] [Revised: 12/15/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Neuroendocrine tumors (NET) of the lung are a slowly growing subtype of lung cancer that has a different treatment paradigm than aggressive and more common forms of lung neuroendocrine neoplasms (NEN) like small cell lung cancer (SCLC). Current guidelines for metastatic lung NET advocate a handful of treatment options, including somatostatin analogs (SSA), everolimus, temozolomide- or platin-based chemotherapy, and peptide receptor radionuclide therapy (PRRT). However, there is no clear treatment sequence, and the therapy of choice may depend on several factors such as tumor grade / growth rate, tumor burden / symptoms, disease progression status, and somatostatin receptor (SSTR) expression. In order to tailor treatment to each individual patient, the latest scientific findings and patient-specific clinical features must be considered together. This review critically evaluates the available evidence with regards to relevant patient characteristics, inclusion and exclusion criteria, and outcome metrics of clinical trials given the presumed natural disease course. Specific patient subgroups with an unmet therapeutic need are identified and discussed in the context of ongoing clinical trials.
Collapse
Affiliation(s)
- Philipp Melhorn
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Austria
| | - Markus Raderer
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Austria
| | - Barbara Kiesewetter
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Austria.
| |
Collapse
|
33
|
Nagashima S, Kobayashi S, Tsunoda S, Yamachika Y, Tozuka Y, Fukushima T, Morimoto M, Ueno M, Furuse J, Maeda S. A case of complete remission by cabozantinib as an end-line treatment for advanced hepatocellular carcinoma. Clin J Gastroenterol 2025; 18:125-129. [PMID: 39616585 PMCID: PMC11785699 DOI: 10.1007/s12328-024-02062-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/07/2024] [Indexed: 02/01/2025]
Abstract
Cabozantinib is a multi-kinase inhibitor targeting multiple tyrosine kinases. It improves overall survival and progression-free survival in patients previously treated with sorafenib for advanced hepatocellular carcinoma (HCC) compared to the placebo in the phase 3 CELESTIAL trial. A 71-year-old man presented to our hospital for treatment of HCC with chronic hepatitis C. He was refractory to sorafenib, lenvatinib, regorafenib, and ramucirumab and started atezolizumab and bevacizumab therapy in November 2020. After administering the second cycle on December 10, 2020, the patient was diagnosed with progressive disease in January 2021. Therefore, cabozantinib (60 mg/day) was initiated on January 14, 2021. As the grade 3 aspartate aminotransferase and alanine aminotransferase levels increased, grade 3 anorexia and a decline in performance status were observed in the first week, and cabozantinib was terminated. His performance status and anorexia gradually improved, and contrast-enhanced computed tomography (CT) in June 2021 showed complete remission (CR) according to the modified Response Evaluation Criteria in Solid Tumors. The patient did not show disease progression for 11 months without receiving any treatment for HCC. To the best of our knowledge, this is the first report of CR with cabozantinib in advanced HCC.
Collapse
Affiliation(s)
- Shuhei Nagashima
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan.
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| | - Satoshi Kobayashi
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Shotaro Tsunoda
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Yui Yamachika
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Yuichiro Tozuka
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Taito Fukushima
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Manabu Morimoto
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao Asahi-ku, Yokohama, 241-8515, Japan
| | - Shin Maeda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| |
Collapse
|
34
|
Abd Elhameed AA, Ali AR, Ghabbour HA, Bayomi SM, El-Gohary NS. Probing structural requirements for thiazole-based mimetics of sunitinib as potent VEGFR-2 inhibitors. RSC Med Chem 2025:d4md00754a. [PMID: 39850549 PMCID: PMC11753467 DOI: 10.1039/d4md00754a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/29/2024] [Indexed: 01/25/2025] Open
Abstract
Novel thiazole analogs 3a, 3b, 4, 5, 6a-g, 8a, 8b, 9a-c, 10a-d and 11 were designed and synthesized as molecular mimetics of sunitinib. In vitro antitumor activity of the obtained compounds was investigated against HepG2, HCT-116, MCF-7, HeP-2 and HeLa cancer cell lines. The obtained data showed that compounds 3b and 10c are the most potent members toward HepG2, HCT-116, MCF-7 and HeLa cells. Moreover, compounds 3a, 3b, 6g, 8a and 10c were assessed for their in vitro VEGFR-2 inhibitory activity. Results proved that compound 10c exhibited outstanding VEGFR-2 inhibition (IC50 = 0.104 μM) compared to sunitinib. Compound 10c paused the G0-G1 phase of the cell cycle in HCT-116 and MCF-7 cells and the S phase in HeLa cells. Additionally, compound 10c elevated caspase-3/9 levels in HCT-116 and HeLa cells, leading to cancer cell death via apoptosis. Furthermore, compound 10c showed a significant reduction in tumor volume in Swiss albino female mice as an in vivo breast cancer model. Docking results confirmed the tight binding interactions of compound 10c with the VEGFR-2 binding site, with its binding energy surpassing that of sunitinib. In silico PK studies predicted compound 10c to have good oral bioavailability and a good drug score with low human toxicity risks.
Collapse
Affiliation(s)
- Alaa A Abd Elhameed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Ahmed R Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Hazem A Ghabbour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Said M Bayomi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Nadia S El-Gohary
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| |
Collapse
|
35
|
Liu X, Lei X, Huang S, Yang X. Current Perspectives of Immunotherapy for Hepatocellular Carcinoma. Comb Chem High Throughput Screen 2025; 28:185-201. [PMID: 38031784 DOI: 10.2174/0113862073255266231025111125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/10/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023]
Abstract
Hepatocellular carcinoma is the sixth most common tumor and the third leading cause of cancer death worldwide. It ranks fourth in the spectrum of malignant tumor incidence and second in the order of death from major malignant tumors in China. Hepatocellular carcinoma is a complex ecosystem containing non-tumor cells (mainly immune-related cells), and its immunotherapy can stimulate the recognition of specific tumor antigens, inhibit the proliferation of cancer cells, and produce over-memory lymphocytes, which can prevent recurrence. So, immunotherapy of hepatocellular carcinoma is increasingly becoming a research hotspot in liver cancer treatment. With the intensive research in recent years, great progress has been made in immunotherapy for hepatocellular carcinoma, including immune checkpoint inhibitors, pericyte therapy, vaccination, and antiviral therapy. In addition, the study found that the therapeutic effect of combination therapy was enhanced compared to monotherapy. This review summarizes the most prominent immunotherapies currently available for the clinical treatment of patients with HCC and the main opportunities and challenges facing HCC research.
Collapse
Affiliation(s)
- Xiaoyi Liu
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, People's Republic of China
| | - Xiaoyong Lei
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, People's Republic of China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, People's Republic of China
| | - Sheng Huang
- Jiuzhitang Co., Ltd, Changsha, Hunan 410007, People's Republic of China
| | - Xiaoyan Yang
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, People's Republic of China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, People's Republic of China
| |
Collapse
|
36
|
Zhao L, Qian C, Ma X, Wang X. Bioactive Products Targeting C-Met As Potential Antitumour Drugs. Anticancer Agents Med Chem 2025; 25:688-696. [PMID: 39812063 DOI: 10.2174/0118715206346207241217064022] [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: 08/12/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 01/16/2025]
Abstract
Mesenchymal‒epithelial transition factor (c-Met), a receptortyrosine kinase (RTK), plays a vital role in cell proliferation, migration and invasion, and tumour metastasis. OBJECTIVE With increasing duration of treatment, many tumours gradually develop drug resistance. Therefore, novel antitumour drugs need to be developed to treat patients with tumours. Targeting c-met inhibitors may be an effective treatment strategy. METHODS Scientific databases such as ScienceDirect, PubMed, the Wiley Online Library, and Social Sciences Citation Index were used to collect information. All the relevant literature was reviewed, and the available literature was screened. The upstream and downstream pathways of c-Met and their relevance to antitumour effects were searched based on the articles' title, abstract, and full text. The c-Met-targeting drugs with antitumour effects are summarized below. A "citation within a citation" or snowballing approach was used in this screening process to identify additional papers that may have been missed in the initial literature screening process. High-quality studies published in peer-reviewed journals were summarized and prioritized for citation in the review. RESULTS In recent years, research on small-molecule targeted drugs has developed rapidly. Many results have also been achieved in the synthesis and isolation of c-Met inhibitors from natural compounds and traditional Chinese medicines. CONCLUSION This article summarizes the developments in anti-c-Met drugs, which are synthesized and isolated from natural compounds and traditional Chinese medicine (TCM). This study provides primary resources for the development of c-Met inhibitors.
Collapse
Affiliation(s)
- Liying Zhao
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chunmei Qian
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xiaoqi Ma
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoyu Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
37
|
Matsumoto D, Naiki T, Naiki‐Ito A, Aoki M, Kato S, Morikawa T, Shimizu N, Gonda M, Umemoto Y, Yasui T. Efficacy of pembrolizumab plus lenvatinib as first-line treatment for metastatic renal cell carcinoma with multiple brain metastases. IJU Case Rep 2025; 8:5-9. [PMID: 39749291 PMCID: PMC11693097 DOI: 10.1002/iju5.12786] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/06/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction Patients with metastatic renal cell carcinoma have a poor prognosis and its specific pathogenesis remains unelucidated. Case presentation At 78 years of age, a Japanese male patient was diagnosed with metastatic renal cell carcinoma (cT3N2M1 stage) and multiple brain metastases that were responsive to stereotactic radiation therapy followed by systemic combination induction therapy of pembrolizumab plus lenvatinib. Adverse events, including grade 3 hypertension, grade 2 eruption, and elevated grade 2 fever, were controlled by a dose reduction or suspension of drugs. The patient eventually showed a tolerance for continuing with 8 mg lenvatinib. Fourteen months after the initiation of treatment, and on 8 mg lenvatinib, this patient showed no sign of disease progression at the last follow-up. Conclusion We detail the absence of disease progression in a metastatic renal cell carcinoma case with multiple brain metastases more than 1 year after stereotactic radiation therapy followed by first-line pembrolizumab plus lenvatinib.
Collapse
Affiliation(s)
- Daisuke Matsumoto
- Department of UrologyNagoya City University West Medical CenterNagoyaJapan
| | - Taku Naiki
- Department of UrologyNagoya City University West Medical CenterNagoyaJapan
- Department of Nephro‐urologyGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Aya Naiki‐Ito
- Department of Experimental Pathology and Tumor BiologyGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Maria Aoki
- Department of UrologyNagoya City University West Medical CenterNagoyaJapan
| | - Shinji Kato
- Department of RadiologyGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Toshiharu Morikawa
- Department of Nephro‐urologyGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Nobuhiko Shimizu
- Department of Nephro‐urologyGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Masakazu Gonda
- Department of Nephro‐urologyGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Yukihiro Umemoto
- Department of UrologyNagoya City University West Medical CenterNagoyaJapan
| | - Takahiro Yasui
- Department of Nephro‐urologyGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| |
Collapse
|
38
|
Dhawale SA, Mokale SN, Dabhade PS. Discovery of Novel Pyrimidine Based Small Molecule Inhibitors as VEGFR-2 Inhibitors: Design, Synthesis, and Anti-cancer Studies. Curr Comput Aided Drug Des 2025; 21:38-49. [PMID: 38185893 DOI: 10.2174/0115734099269413231018065351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/02/2023] [Accepted: 09/12/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Receptor tyrosine kinases (RTKs) are potent oncoproteins in cancer that, when mutated or overexpressed, can cause uncontrolled growth of cells, angiogenesis, and metastasis, making them significant targets for cancer treatment. Vascular endothelial growth factor receptor 2 (VEGFR2), is a tyrosine kinase receptor that is produced in endothelial cells and is the most crucial regulator of angiogenic factors involved in tumor angiogenesis. So, a series of new substituted N-(4-((2-aminopyrimidin-5-yl)oxy)phenyl)-N-phenyl cyclopropane- 1,1-dicarboxamide derivatives as VEGFR-2 inhibitors have been designed and synthesized. METHODS Utilizing H-NMR, C13-NMR, and mass spectroscopy, the proposed derivatives were produced and assessed. HT-29 and COLO-205 cell lines were used for the cytotoxicity tests. The effective compound was investigated further for the Vegfr-2 kinase inhibition assay, cell cycle arrest, and apoptosis. A molecular docking examination was also carried out with the Maestro-12.5v of Schrodinger. RESULTS In comparison to the reference drug Cabozantinib (IC50 = 9.10 and 10.66 μM), compound SP2 revealed promising cytotoxic activity (IC50 = 4.07 and 4.98 μM) against HT-29 and COLO-205, respectively. The synthesized compound SP2 showed VEGFR-2 kinase inhibition activity with (IC50 = 6.82 μM) against the reference drug, Cabozantinib (IC50 = 0.045 μM). Moreover, compound SP2 strongly induced apoptosis by arresting the cell cycle in the G1 phase. The new compounds' potent VEGFR-2 inhibitory effect was noted with key amino acids Asp1044, and Glu883, and the hydrophobic interaction was also observed in the pocket of the VEGFR-2 active site by using a docking study. CONCLUSION The results demonstrate that at the cellular and enzyme levels, the synthetic compounds SP2 are similarly effective as cabozantinib. The cell cycle and apoptosis data demonstrate the effectiveness of the suggested compounds. Based on the findings of docking studies, cytotoxic effects, in vitro VEGFR-2 inhibition, apoptosis, and cell cycle arrest, this research has given us identical or more effective VEGFR-2 inhibitors.
Collapse
Affiliation(s)
- Sachin A Dhawale
- Department of Pharmaceutical Chemistry, Y.B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, 431001, Maharashtra, India
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, 431001, M.S. India
| | - Santosh N Mokale
- Department of Pharmaceutical Chemistry, Y.B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, 431001, Maharashtra, India
| | - Pratap S Dabhade
- Department of Pharmaceutical Chemistry, Y.B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, 431001, Maharashtra, India
| |
Collapse
|
39
|
Chen A, Yin K, Liu Y, Hu L, Cui Q, Wan X, Yang W. WEE Family Kinase Inhibitors Combined with Sorafenib Can Selectively Inhibit HCC Cell Proliferation. Curr Cancer Drug Targets 2025; 25:370-385. [PMID: 38860904 DOI: 10.2174/0115680096298370240520093003] [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: 01/08/2024] [Revised: 04/10/2024] [Accepted: 04/24/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Sorafenib is currently the first choice for the treatment of patients with advanced hepatocellular carcinoma, but its therapeutic effect is still limited. OBJECTIVES This study aims to examine whether WEE family kinase inhibitors can enhance the anticancer effect of sorafenib. METHODS We analyzed the expression levels of PKMYT1 kinase and WEE1 kinase in HCC, studied the inhibitory effect of PKMYT1 kinase inhibitor RP-6306, WEE1 kinase inhibitor adavosertib combined with sorafenib on the proliferation of HCC cells, and detected the effect of drug combination on CDK1 phosphorylation. RESULTS We found that PKMYT1 and WEE1 were upregulated in HCC and were detrimental to patient survival. Cell experiments showed that both RP-6306 and adavosertib (1-100 μM) inhibited the proliferation of HCC cell lines in a dose-dependent manner alone, and the combination of the two drugs had a synergistic effect. In HCC cell lines, sorafenib combined with RP-6306 or adavosertib showed a synergistic antiproliferation effect and less toxicity to normal cells. Sorafenib combined with RP-6306 and adavosertib further inhibited the proliferation of HCC cells and caused complete dephosphorylation of CDK1. CONCLUSION Taken together, our findings provide experimental evidence for the future use of sorafenib in combination with RP-6306 or adavosertib for the treatment of HCC.
Collapse
Affiliation(s)
- Anling Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230031, China
| | - Ke Yin
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yu Liu
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Lei Hu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Qianwen Cui
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230031, China
| | - Xiaofeng Wan
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| |
Collapse
|
40
|
Patil J, Bhattacharya S, Saoji SD, Dande P. Cabozantinib-phospholipid complex for enhanced solubility, bioavailability, and reduced toxicity in liver cancer. Ther Deliv 2025; 16:25-41. [PMID: 39611708 PMCID: PMC11703380 DOI: 10.1080/20415990.2024.2435240] [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: 11/03/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
AIMS To enhance the therapeutic potential of Cabozantinib (CBZ), a tyrosine kinase inhibitor with limited water solubility, low bioavailability, and high toxicity, by developing a Cabozantinib-Phospholipid Complex (CBZ-PLS). MATERIALS & METHODS CBZ-PLS was formulated using solvent evaporation with a Box-Behnken design and characterized using various techniques to confirm molecular interactions. Solubility, in vitro release, pharmacokinetics, and toxicity were evaluated. Cytotoxic effects on HepG2 cell lines were also assessed. RESULTS CBZ-PLS exhibited a 126-fold increase in solubility and enhanced CBZ release in vitro. Pharmacokinetic studies on Wistar rats demonstrated a 1.58-fold increase in bioavailability, while acute toxicity studies confirmed biocompatibility. CBZ-PLS showed superior cytotoxicity, apoptosis induction, migration inhibition, increased ROS generation, and greater DNA fragmentation in HepG2 cells. The complex also maintained stability over 6 months. CONCLUSIONS CBZ-PLS significantly improves the solubility, bioavailability, and therapeutic efficacy of CBZ against liver cancer, presenting a promising approach for more effective liver cancer treatment.
Collapse
Affiliation(s)
- Jayesh Patil
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM’S NMIMS Deemed-to-be University, Shirpur, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM’S NMIMS Deemed-to-be University, Shirpur, India
| | - Suprit D. Saoji
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University Nagpur, Mahatma Jyotiba Fuley Shaikshanik Parisar, University Campus, Nagpur, India
| | - Payal Dande
- Department of Pharmacognosy, School of Pharmacy & Technology Management, SVKM’S NMIMS Deemed-to-be University, Shirpur, India
| |
Collapse
|
41
|
Haga Y, Ray R, Ray RB. Silmitasertib in Combination With Cabozantinib Impairs Liver Cancer Cell Cycle Progression, Induces Apoptosis, and Delays Tumor Growth in a Preclinical Model. Mol Carcinog 2025; 64:72-82. [PMID: 39377735 DOI: 10.1002/mc.23827] [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: 08/06/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024]
Abstract
The rising incidence of hepatocellular carcinoma (HCC) is a global problem. Several approved treatments, including immune therapy and multi-tyrosine kinase inhibitors, are used for treatment, although the results are not optimum. There is an unmet need to develop highly effective chemotherapies for HCC. Targeting multiple pathways to attack cancer cells is beneficial. Cabozantinib is an orally available bioactive multikinase inhibitor and has a modest effect on HCC treatment. Silmitasertib is an orally bioavailable, potent CK2 inhibitor with a direct role in DNA damage repair and is in clinical trials for other cancers. In this study, we planned to repurpose these existing drugs on HCC treatment. We observed a stronger antiproliferative effect of these two combined drugs on HCC cells generated from different etiologies as compared to the single treatment. Global RNA-seq analyses revealed a decrease in the expression of G2/M cell cycle transition genes in HCC cells following combination treatment, suggesting G2 phase cell arrest. We observed G2/M cell cycle phase arrest in HCC cells upon combination treatment compared to the single-treated or vehicle-treated control cells. The downregulation of CCNA2 and CDC25C following combination therapy further supported the observation. Subsequent analyses demonstrated that combination treatment inhibited 70 kDa ribosomal protein S6 kinase (p70S6K) phosphorylation, and increased Bim expression. Apoptosis of HCC cells were accompanied by increased poly (ADP-ribose) polymerase cleavage and caspase-9 activation. Next, we observed that a combination therapy significantly delayed the progression of HCC xenograft growth as compared to vehicle control. Together, our results suggested combining cabozantinib and silmitasertib would be a promising treatment option for HCC.
Collapse
Affiliation(s)
- Yuki Haga
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, USA
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, Missouri, USA
| | - Ratna B Ray
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, Missouri, USA
- Department of Pathology, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
42
|
Hu W, Peng X, Ji Y, Duan W, Ai J, Zhan Z. Incorporation of a rigid 1,3-diketone-containing fragment led to significantly improved AXL inhibitory activity: design, synthesis, and SAR of the anilinopyrimidine AXL inhibitors. Mol Divers 2024:10.1007/s11030-024-11071-9. [PMID: 39731692 DOI: 10.1007/s11030-024-11071-9] [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: 10/25/2024] [Accepted: 11/25/2024] [Indexed: 12/30/2024]
Abstract
Overexpressed AXL kinase is involved in various human malignancies, which incurs tumor progression, poor prognosis, and drug resistance. Suppression of the aberrant AXL axis with genetic tools or small-molecule inhibitors has achieved valid antitumor efficacies in both preclinical studies and clinical antitumor campaigns. Herein we will report the design, synthesis, and structure-activity relationship (SAR) exploration of a series of anilinopyrimidine type II AXL inhibitors. Among these inhibitors, 4l exhibited the enzymatic AXL and cellular BaF3/TEL-AXL IC50 values of 0.5 nM and less than 0.2 nM, respectively. Western blot analysis displayed that 4l dose-dependently inhibited the phosphorylation of AXL and its downstream cascade Akt, which was better than that of the reference control R428. Moreover, 4l markedly suppressed the AXL/GAS6-mediated migration in NCI-H1299 cells.
Collapse
Affiliation(s)
- Wenyi Hu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Xia Peng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, Jiangsu, People's Republic of China
| | - Yinchun Ji
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, Jiangsu, People's Republic of China
| | - Wenhu Duan
- Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, People's Republic of China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, People's Republic of China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 1000449, People's Republic of China
| | - Jing Ai
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, Jiangsu, People's Republic of China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 1000449, People's Republic of China.
| | - Zhengsheng Zhan
- Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, People's Republic of China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, People's Republic of China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 1000449, People's Republic of China.
| |
Collapse
|
43
|
Mormando M, Lauretta R, Puliani G, Bianchini M, Spoltore ME, Appetecchia M. Treatment Outcomes and Toxicities of Multiple Tyrosin Kinase Inhibitors for Metastatic Medullary Thyroid Cancer: A Case Series. Biomedicines 2024; 12:2923. [PMID: 39767829 PMCID: PMC11673415 DOI: 10.3390/biomedicines12122923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The current possible treatments of advanced medullary carcinoma (MTC) include different drugs belonging to the class of tyrosine kinase inhibitors (TKIs): vandetanib, cabozantinb, and selpercatinib. Although the effects of these TKIs have been well described in clinical trials, the real-practice evidence of the effectiveness and safety of these treatment is scant. This real-world case series aims to describe a niche of patients with advanced MTC treated with more than one TKI by focusing on treatment responses and any reported adverse events (AEs) and to provide additional insight on the individualized approach to the management of metastatic MTC. Methods: Five patients with a diagnosis of metastastic MTC, treated with at least two different molecules of TKIs, were retrospectively selected. Results: Three patients obtained a partial response (one with cabozantinb, one with selpercatinib, and one with vandetanib), and two patients obtained disease stability (both of them treated with all three TKIs, the first two lines discontinued for AEs). The AE profile agreed with the known clinical trials AEs except for non-neoplastic ascites related to selpercatinib and lung cavitations of non-neoplastic tissue related to cabozantinb. The latter was an AE never described so far in patients receiving TKIs. Conclusions: The best management of MTC relies on an individualized approach, keeping in mind and dealing with the potential toxicity in order to minimize the treatment withdrawal.
Collapse
Affiliation(s)
- Marilda Mormando
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (M.M.); (R.L.); (G.P.); (M.B.)
| | - Rosa Lauretta
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (M.M.); (R.L.); (G.P.); (M.B.)
| | - Giulia Puliani
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (M.M.); (R.L.); (G.P.); (M.B.)
| | - Marta Bianchini
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (M.M.); (R.L.); (G.P.); (M.B.)
| | - Maria Elena Spoltore
- Section of Medical Pathophysiology, Food Science and Endocrinology, Department of Experimental Medicine, Sapienza University of Roma, 00185 Rome, Italy;
| | - Marialuisa Appetecchia
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (M.M.); (R.L.); (G.P.); (M.B.)
| |
Collapse
|
44
|
Lutimba S, Saleem B, Aleem E, Mansour MA. In Silico Analysis of Triamterene as a Potential Dual Inhibitor of VEGFR-2 and c-Met Receptors. J Xenobiot 2024; 14:1962-1987. [PMID: 39728413 DOI: 10.3390/jox14040105] [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: 11/19/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
The vascular endothelial growth factor receptor 2 (VEGFR2) and the hepatocyte growth factor receptor (C-Met) are critical receptors for signaling pathways controlling crucial cellular processes such as cell growth, angiogenesis and tissue regeneration. However, dysregulation of these proteins has been reported in different diseases, particularly cancer, where these proteins promote tumour growth, invasiveness, metastasis and resistance to conventional therapies. The identification of dual inhibitors targeting both VEGFR-2 and c-Met has emerged as a strategic therapeutic approach to overcome the limitations and resistance mechanisms associated with single-target therapies in clinical settings. Through molecular dynamics simulations and comparative docking analysis, we tested the inhibitory potential of 2,016 Food and Drug Administration (FDA)-approved drugs targeting VEGFR-2 and/or c-Met receptors. The results revealed that entacapone and telmisartan are potent and selective inhibitors for c-Met and VEGFR-2, respectively. Interestingly, triamterene was identified as a promising dual inhibitor, demonstrating specific and significant binding affinity to both proteins. Molecular dynamics simulations revealed key interactions between the identified compounds and critical residues in the catalytic domains of both VEGFR-2 (e.g., Lys868, Asp1028, Asp1046) and c-Met (e.g., Asp1204, His1202, Asp1222), providing insights into their mechanism of action. These findings underscore the therapeutic potential of triamterene in targeting multiple signaling pathways involved in cancer progression, metastasis and poor prognosis in patients. Our study provides a foundational framework for the development of novel anticancer compounds able to target multiple pathways in cancer. Further preclinical and clinical investigations are needed to validate the efficacy of these compounds in clinical settings and to test their ability to overcome resistance and improve patient outcome.
Collapse
Affiliation(s)
- Stuart Lutimba
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
| | - Baraya Saleem
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
| | - Eiman Aleem
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
| | - Mohammed A Mansour
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
45
|
Carra S, Gaudenzi G, Franceschetti G, Collini M, Sironi L, Bouzin M, Persani L, Chirico G, Vitale G, D’Alfonso L. How Tumors Affect Hemodynamics: A Diffusion Study on the Zebrafish Transplantable Model of Medullary Thyroid Carcinoma by Selective Plane Illumination Microscopy. Int J Mol Sci 2024; 25:13392. [PMID: 39769158 PMCID: PMC11678154 DOI: 10.3390/ijms252413392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Medullary thyroid carcinoma (MTC), a rare neuroendocrine tumor comprising 3-5% of thyroid cancers, arises from calcitonin-producing parafollicular C cells. Despite aggressive behavior, surgery remains the primary curative treatment, with limited efficacy reported for radiotherapy and chemotherapy. Recent efforts have explored the pathogenetic mechanisms of MTC, identifying it as a highly vascularized neoplasm overexpressing pro-angiogenic factors. Building on the established benefits of zebrafish embryos, we previously created an in vivo MTC xenograft platform that allows real-time observation of tumor-induced angiogenesis and evaluation of the anti-angiogenic effects of tyrosine kinase inhibitors. In this study, we present a method using selective plane illumination microscopy (SPIM) to characterize vascular permeability in these xenografted embryos. Taking advantage of dextran injections into the blood flow of zebrafish embryos, we found that the diffusion coefficient in embryos grafted with MTC cells was about tenfold lower compared with the same parameter in controls. The results demonstrate the potential of our approach to estimate diffusion parameters, providing valuable insights into vascular permeability changes in MTC-implanted zebrafish embryos compared with controls. Our study sheds light on the intricate vascular biology of MTC, offering a promising tool for future investigations into tumor-induced angiogenesis and therapeutic strategies in diverse neoplasms.
Collapse
Affiliation(s)
- Silvia Carra
- Laboratory of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, 20100 Milan, Italy;
| | - Germano Gaudenzi
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, IRCCS Istituto Auxologico Italiano, 20100 Milan, Italy; (G.G.); (G.V.)
| | - Giorgia Franceschetti
- Department of Physics “G. Occhialini”, Università degli Studi di Milano-Bicocca, Piazza Della Scienza 3, 20126 Milan, Italy (M.C.); (L.S.); (M.B.); (G.C.); (L.D.)
| | - Maddalena Collini
- Department of Physics “G. Occhialini”, Università degli Studi di Milano-Bicocca, Piazza Della Scienza 3, 20126 Milan, Italy (M.C.); (L.S.); (M.B.); (G.C.); (L.D.)
| | - Laura Sironi
- Department of Physics “G. Occhialini”, Università degli Studi di Milano-Bicocca, Piazza Della Scienza 3, 20126 Milan, Italy (M.C.); (L.S.); (M.B.); (G.C.); (L.D.)
| | - Margaux Bouzin
- Department of Physics “G. Occhialini”, Università degli Studi di Milano-Bicocca, Piazza Della Scienza 3, 20126 Milan, Italy (M.C.); (L.S.); (M.B.); (G.C.); (L.D.)
| | - Luca Persani
- Laboratory of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, 20100 Milan, Italy;
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20100 Milan, Italy
| | - Giuseppe Chirico
- Department of Physics “G. Occhialini”, Università degli Studi di Milano-Bicocca, Piazza Della Scienza 3, 20126 Milan, Italy (M.C.); (L.S.); (M.B.); (G.C.); (L.D.)
| | - Giovanni Vitale
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, IRCCS Istituto Auxologico Italiano, 20100 Milan, Italy; (G.G.); (G.V.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20100 Milan, Italy
| | - Laura D’Alfonso
- Department of Physics “G. Occhialini”, Università degli Studi di Milano-Bicocca, Piazza Della Scienza 3, 20126 Milan, Italy (M.C.); (L.S.); (M.B.); (G.C.); (L.D.)
| |
Collapse
|
46
|
Wu J, Mo H, An Z, Tang Z, Deng X, Zhou H, Gong Y, Zheng C, Zhuo L, Tan S. Discovery of 7-(1-methyl-1H-pyrazol-4-yl)-1,6-naphthyridine derivatives as potent inhibitors of rearranged during transfection (RET) and RET solvent-front mutants for overcoming selpercatinib resistance. Eur J Med Chem 2024; 279:116891. [PMID: 39316846 DOI: 10.1016/j.ejmech.2024.116891] [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: 07/31/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Rearranged during transfection kinase (RET) inhibition has been considered a promising therapeutic approach for treatment of a variety of cancers. However, the clinical therapeutic benefits of the second-generation RET inhibitor selpercatinib are greatly compromised by acquired resistance mediated by solvent-front mutations (e.g., RETG810 R/S/C). Herein, we report a class of 7-(1-methyl-1H-pyrazol-4-yl)-1,6-naphthyridine derivatives as potent RET and RET solvent-front mutant inhibitors for overcoming selpercatinib resistance. The representative compound 20p exhibited excellent in vitro inhibitory activities against solvent-front mutations (RETG810R, RETG810S, and RETG810C) with low nanomolar range (IC50 of 5.7-8.3 nM), which was 15-29-fold more potent than selpercatinib (IC50 of 95.3-244.1 nM). Additionally, 20p exhibited acceptable pharmacokinetic properties with oral bioavailability of 30.4 %. Importantly, 20p exhibited highly impressive antitumor potency in both a Ba/F3-KIF5B-RETWT-derived xenograft mouse model and a selpercatinib-resistant Ba/F3-KIF5B-RETG810R-positive mutant xenograft mouse model. Overall, 20p represents a novel and promising drug lead for overcoming RET solvent-front mutation-based resistance to selpercatinib.
Collapse
Affiliation(s)
- Junbo Wu
- Department of Colorectal Surgery, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, 421001, Hunan, China
| | - Hanxuan Mo
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhigang An
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zishu Tang
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xinyu Deng
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Huifang Zhou
- Department of Colorectal Surgery, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, 421001, Hunan, China
| | - Yi Gong
- National Key Laboratory of Green Pesticide, Central China Normal University, Wuhan, 430079, China
| | - Chenggong Zheng
- Pulmonary Hospital, Changsha Central Hospital, Changsha, Hunan, 410004, China
| | - Linsheng Zhuo
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Shuguang Tan
- Department of Colorectal Surgery, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, 421001, Hunan, China.
| |
Collapse
|
47
|
Smith EA, Belote RL, Cruz NM, Moustafa TE, Becker CA, Jiang A, Alizada S, Prokofyeva A, Chan TY, Seasor TA, Balatico M, Cortes-Sanchez E, Lum DH, Hyngstrom JR, Zeng H, Deacon DC, Grossmann AH, White RM, Zangle TA, Judson-Torres RL. Receptor tyrosine kinase inhibition leads to regression of acral melanoma by targeting the tumor microenvironment. J Exp Clin Cancer Res 2024; 43:317. [PMID: 39627834 PMCID: PMC11613472 DOI: 10.1186/s13046-024-03234-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/13/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Acral melanoma (AM) is an aggressive melanoma variant that arises from palmar, plantar, and nail unit melanocytes. Compared to non-acral cutaneous melanoma (CM), AM is biologically distinct, has an equal incidence across genetic ancestries, typically presents in advanced stage disease, is less responsive to therapy, and has an overall worse prognosis. METHODS An independent analysis of published sequencing data was performed to evaluate the frequency of receptor tyrosine kinase (RTK) ligands and adapter protein gene variants and expression. To target these genetic variants, a zebrafish acral melanoma model and preclinical patient-derived xenograft (PDX) mouse models were treated with a panel of RTK inhibitors. Residual PDX tumors were evaluated for changes in proliferation, vasculature, necrosis, and ferroptosis by histology and immunohistochemistry. RESULTS RTK ligands and adapter proteins are frequently amplified, translocated, and/or overexpressed in AM. Dual FGFR/VEGFR inhibitors decrease acral-analogous melanocyte proliferation and migration in zebrafish, and the potent pan-FGFR/VEGFR inhibitor, Lenvatinib, uniformly induces tumor regression in AM PDX tumors but only slows tumor growth in CM models. Unlike other multi-RTK inhibitors, Lenvatinib is not directly cytotoxic to dissociated AM PDX tumor cells and instead disrupts tumor architecture and vascular networks. CONCLUSION Considering the great difficulty in establishing AM cell culture lines, these findings suggest that AM may be more sensitive to microenvironment perturbations than CM. In conclusion, dual FGFR/VEGFR inhibition may be a viable therapeutic strategy that targets the unique biology of AM.
Collapse
Affiliation(s)
- Eric A Smith
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Rachel L Belote
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
| | - Nelly M Cruz
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tarek E Moustafa
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Carly A Becker
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA
| | - Amanda Jiang
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Shukran Alizada
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA
| | | | - Tsz Yin Chan
- Preclinical Research Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Tori A Seasor
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Michael Balatico
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Emilio Cortes-Sanchez
- Immuno Oncology Network Core, The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - David H Lum
- Preclinical Research Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - John R Hyngstrom
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hanlin Zeng
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dekker C Deacon
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA
| | - Allie H Grossmann
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Richard M White
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Nuffield Department of Medicine, Ludwig Cancer Research, University of Oxford, Oxford, UK
| | - Thomas A Zangle
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Robert L Judson-Torres
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA.
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
48
|
Myall NJ, Das M. ROS1-rearranged non-small cell lung cancer: Understanding biology and optimizing management in the era of new approvals. Curr Probl Cancer 2024; 53:101133. [PMID: 39260124 DOI: 10.1016/j.currproblcancer.2024.101133] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 09/13/2024]
Abstract
Rearrangements involving the ROS1 gene are infrequent in non-small cell lung cancer (NSCLC) but represent an important targetable driver alteration. Occurring most commonly in patients with adenocarcinoma who have a light or never smoking history, ROS1 rearrangements can be identified by either fluorescence in-situ hybridization (FISH) or next-generation sequencing techniques. Multiple tyrosine kinase inhibitors (TKIs) are now available for the effective treatment of ROS1-rearranged NSCLC in the metastatic setting including crizotinib, entrectinib, and repotrectinib as first-line therapy options. In addition, newer targeted therapies with increased selectivity for ROS1 over other targets are also emerging. As treatment of the disease continues to evolve, understanding the clinical course of patients with ROS1-rearranged NSCLC as well as the data supporting the latest therapy options is key to timely, effective, and longitudinal care.
Collapse
Affiliation(s)
- Nathaniel J Myall
- Division of Oncology, Department of Medicine, Stanford Cancer Center, Stanford CA, United States
| | - Millie Das
- Division of Oncology, Department of Medicine, Stanford Cancer Center, Stanford CA, United States; Department of Medicine, VA Palo Alto Health Care System, 3801 Miranda Ave. (111ONC), Palo Alto CA 94304, United States.
| |
Collapse
|
49
|
Gawi Ermi A, Sarkar D. Resistance to Tyrosine Kinase Inhibitors in Hepatocellular Carcinoma (HCC): Clinical Implications and Potential Strategies to Overcome the Resistance. Cancers (Basel) 2024; 16:3944. [PMID: 39682130 DOI: 10.3390/cancers16233944] [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: 10/07/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, and the development of effective treatment strategies remains a significant challenge in the management of advanced HCC patients. The emergence of tyrosine kinase inhibitors (TKIs) has been a significant advancement in the treatment of HCC, as these targeted therapies have shown promise in prolonging the survival of patients with advanced disease. Although immunotherapy is currently considered as the first line of treatment for advanced HCC patients, many such patients do not meet the clinical criteria to be eligible for immunotherapy, and in many parts of the world there is still lack of accessibility to immunotherapy. As such, TKIs still serve as the first line of treatment and play a major role in the treatment repertoire for advanced HCC patients. However, the development of resistance to these agents is a major obstacle that must be overcome. In this review, we explore the underlying mechanisms of resistance to TKIs in HCC, the clinical implications of this resistance, and the potential strategies to overcome or prevent the emergence of resistance.
Collapse
Affiliation(s)
- Ali Gawi Ermi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
50
|
Liao C, Hu L, Zhang Q. Von Hippel-Lindau protein signalling in clear cell renal cell carcinoma. Nat Rev Urol 2024; 21:662-675. [PMID: 38698165 DOI: 10.1038/s41585-024-00876-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
The distinct pathological and molecular features of kidney cancer in adaptation to oxygen homeostasis render this malignancy an attractive model for investigating hypoxia signalling and potentially developing potent targeted therapies. Hypoxia signalling has a pivotal role in kidney cancer, particularly within the most prevalent subtype, known as renal cell carcinoma (RCC). Hypoxia promotes various crucial pathological processes, such as hypoxia-inducible factor (HIF) activation, angiogenesis, proliferation, metabolic reprogramming and drug resistance, all of which contribute to kidney cancer development, growth or metastasis formation. A substantial portion of kidney cancers, in particular clear cell RCC (ccRCC), are characterized by a loss of function of Von Hippel-Lindau tumour suppressor (VHL), leading to the accumulation of HIF proteins, especially HIF2α, a crucial driver of ccRCC. Thus, therapeutic strategies targeting pVHL-HIF signalling have been explored in ccRCC, culminating in the successful development of HIF2α-specific antagonists such as belzutifan (PT2977), an FDA-approved drug to treat VHL-associated diseases including advanced-stage ccRCC. An increased understanding of hypoxia signalling in kidney cancer came from the discovery of novel VHL protein (pVHL) targets, and mechanisms of synthetic lethality with VHL mutations. These breakthroughs can pave the way for the development of innovative and potent combination therapies in kidney cancer.
Collapse
Affiliation(s)
- Chengheng Liao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lianxin Hu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Qing Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|