1
|
Wang C, Liu H, Yang Y, Sun Q, Yin L, Yang L, Wang X, Zhao W, Wan Q, Liu G, Chen Y, Li Z, Wang L. Preliminary Study of Radionuclide-Labeled MerTK-Targeting PET Imaging Agents for the Diagnosis of Melanoma. J Med Chem 2024. [PMID: 39484831 DOI: 10.1021/acs.jmedchem.4c02156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
MerTK PET imaging holds potential as a promising approach for assessing tumor aggressiveness and monitoring treatment response. In this study, we synthesized a series of 18F- and 68Ga-labeled tracers derived from MerTK inhibitors for detection of MerTK expression. Among the synthesized agents, the dimeric compounds [68Ga]10 and [68Ga]12 demonstrated good in vivo and in vitro stability, high affinities to the MerTK receptor, and good MerTK-targeting specificity. Notably, [68Ga]10 exhibited a tumor uptake of 2.6 ± 0.2%ID/g at 1 h p. i. in B16F10 tumor-bearing mice, nearly tripling the uptake of its monomeric counterpart [68Ga]3. A similar enhancement was observed with [68Ga]12 compared to its monomeric analog [68Ga]6. Additionally, [18F]14 achieved a tumor uptake of 7.6 ± 0.5%ID/g at 2 h p. i., outperforming the previously reported [18F]15. Biodistribution analysis further validated the results, highlighting their potential for clinical investigation.
Collapse
Affiliation(s)
- Changjiang Wang
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Hao Liu
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yunyi Yang
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qinghong Sun
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Liping Yin
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Liping Yang
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaodong Wang
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Weiling Zhao
- Department of Radiology, Lineberger Comprehensive Cancer Center, and Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Qiang Wan
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Guangfu Liu
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Yue Chen
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Zibo Li
- Department of Radiology, Lineberger Comprehensive Cancer Center, and Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Li Wang
- Department of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping St, Jiangyang District, Luzhou, Sichuan 646000, China
| |
Collapse
|
2
|
Giordano G, Tucciarello C, Merlini A, Cutrupi S, Pignochino Y. Targeting the EphA2 pathway: could it be the way for bone sarcomas? Cell Commun Signal 2024; 22:433. [PMID: 39252029 PMCID: PMC11382444 DOI: 10.1186/s12964-024-01811-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
Bone sarcomas are malignant tumors of mesenchymal origin. Complete surgical resection is the cornerstone of multidisciplinary treatment. However, advanced, unresectable forms remain incurable. A crucial step towards addressing this challenge involves comprehending the molecular mechanisms underpinning tumor progression and metastasis, laying the groundwork for innovative precision medicine-based interventions. We previously showed that tyrosine kinase receptor Ephrin Type-A Receptor 2 (EphA2) is overexpressed in bone sarcomas. EphA2 is a key oncofetal protein implicated in metastasis, self-renewal, and chemoresistance. Molecular, genetic, biochemical, and pharmacological approaches have been developed to target EphA2 and its signaling pathway aiming to interfere with its tumor-promoting effects or as a carrier for drug delivery. This review synthesizes the main functions of EphA2 and their relevance in bone sarcomas, providing strategies devised to leverage this receptor for diagnostic and therapeutic purposes, with a focus on its applicability in the three most common bone sarcoma histotypes: osteosarcoma, chondrosarcoma, and Ewing sarcoma.
Collapse
Affiliation(s)
- Giorgia Giordano
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Oncology, University of Turin, 10043, Orbassano, TO, Italy
| | - Cristina Tucciarello
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy
| | - Alessandra Merlini
- Department of Oncology, University of Turin, 10043, Orbassano, TO, Italy
| | - Santina Cutrupi
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy
| | - Ymera Pignochino
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy.
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy.
| |
Collapse
|
3
|
Li J, Bai Y, Chen Z, Ying J, Guo Y, Fang W, Zhang F, Xiong J, Zhang T, Meng Z, Zhang J, Ren Z, Hao C, Chen Y, Lin X, Pan H, Zhou F, Li X, Yu F, Zhang J, Zhang Z, Qin S. SAFFRON-104: a phase Ib/II study of sitravatinib alone or with tislelizumab in advanced hepatocellular carcinoma and gastric cancer/gastroesophageal junction cancer. Cancer Immunol Immunother 2024; 73:219. [PMID: 39235596 PMCID: PMC11377389 DOI: 10.1007/s00262-024-03806-2] [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: 04/29/2024] [Accepted: 08/08/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Sitravatinib is a spectrum-selective tyrosine kinase inhibitor targeting TAM (TYRO3, AXL, MER), VEGFR-2, KIT, and MET. SAFFRON-104 (NCT03941873) was a multicohort phase Ib/II study investigating sitravatinib with/without tislelizumab, an anti-programmed cell death protein 1 (PD-1) antibody, in patients with advanced hepatocellular carcinoma (HCC) or gastric cancer/gastroesophageal junction cancer (GC/GEJC). METHODS Eligible patients had histologically/cytologically confirmed advanced HCC or GC/GEJC. Phase I determined the recommended phase II dose (RP2D) of sitravatinib with/without tislelizumab. Phase II evaluated sitravatinib monotherapy in patients with pretreated HCC, and sitravatinib plus tislelizumab in anti-PD-(L)1-naïve or -treated HCC and anti-PD-(L)1-naïve GC/GEJC. Primary endpoints were safety/tolerability (phase I) and objective response rate (ORR) (phase II). RESULTS At data cutoff (March 31, 2023), 111 patients were enrolled; 102 were efficacy-evaluable (median study follow-up 9.1 months [range: 0.7-36.9]). The RP2D of sitravatinib was determined as 120 mg orally once daily. In patients receiving sitravatinib monotherapy and sitravatinib in combination with tislelizumab, grade ≥ 3 treatment-related adverse events occurred in 14 (51.9%) and 42 (50.0%) patients, respectively. The ORR was 25% (95% confidence interval [CI]: 8.7-49.1) in patients with pretreated HCC receiving sitravatinib monotherapy. In patients receiving sitravatinib with tislelizumab, the ORR was 11.5% (95% CI 2.4-30.2) with anti-PD-(L)1-naïve HCC, 9.5% (95% CI 1.2-30.4) with anti-PD-(L)1-treated HCC, and 16.1% (95% CI 5.5-33.7) in patients with anti-PD-(L)1-naïve GC/GEJC. CONCLUSIONS Sitravatinib with/without tislelizumab was generally well tolerated and showed preliminary antitumor activity in patients with advanced HCC and GC/GEJC.
Collapse
Affiliation(s)
- Jin Li
- Department of Oncology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yuxian Bai
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhendong Chen
- Department of Oncology, The Second Hospital of Anhui Medical Hospital, Hefei, China
| | - Jieer Ying
- Department of Hepato-Pancreato-Biliary and Gastric Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yabing Guo
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Zhang
- Department of Hepatic & Biliary & Pancreatic Surgery, Hubei Cancer Hospital, Wuhan, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Zhang
- Department of Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Meng
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jingdong Zhang
- Medical Oncology Department of Gastrointestinal Cancer, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Zhenggang Ren
- Department of Hepatic Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunyi Hao
- Department of Hepato-Pancreato-Biliary Surgery, Beijing Cancer Hospital, Beijing, China
| | - Yajin Chen
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyan Lin
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fuxiang Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xin Li
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Fan Yu
- BeiGene (Shanghai) Co., Ltd., Shanghai, China
| | - Juan Zhang
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | | | - Shukui Qin
- GI Cancer Center, Nanjing Tianyinshan Hospital of China Pharmaceutical University, Gulou, Nanjing, Jiangsu, China.
| |
Collapse
|
4
|
Vázquez-Bellón N, Martínez-Bosch N, García de Frutos P, Navarro P. Hallmarks of pancreatic cancer: spotlight on TAM receptors. EBioMedicine 2024; 107:105278. [PMID: 39137571 PMCID: PMC11367522 DOI: 10.1016/j.ebiom.2024.105278] [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: 04/19/2024] [Revised: 07/14/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents the most prevalent type of pancreatic cancer and ranks among the most aggressive tumours, with a 5-year survival rate of less than 11%. Projections indicate that by 2030, it will become the second leading cause of cancer-related deaths. PDAC presents distinctive hallmarks contributing to its dismal prognosis: (i) late diagnosis, (ii) heterogenous and complex mutational landscape, (iii) high metastatic potential, (iv) dense fibrotic stroma, (v) immunosuppressive microenvironment, and (vi) high resistance to therapy. Mounting evidence has shown a role for TAM (Tyro3, AXL, MerTK) family of tyrosine kinase receptors in PDAC initiation and progression. This review aims to describe the impact of TAM receptors on the defining hallmarks of PDAC and discuss potential future directions using these proteins as novel biomarkers for early diagnosis and targets for precision therapy in PDAC, an urgent unmet clinical need.
Collapse
Affiliation(s)
- Núria Vázquez-Bellón
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB)-CSIC and Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain; PhD Program in Biomedicine, Facultat de Medicina (Campus Clínic), Universitat de Barcelona, Barcelona, Spain
| | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Research Institute (HMRI), Unidad Asociada IIBB-CSIC, Barcelona, Spain
| | - Pablo García de Frutos
- Department of Cell Death and Proliferation, IIBB-CSIC, Unidad Asociada IMIM/IIBB-CSIC, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), and IDIBAPS, Barcelona, Spain.
| | - Pilar Navarro
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB)-CSIC and Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain; Cancer Research Program, Hospital del Mar Research Institute (HMRI), Unidad Asociada IIBB-CSIC, Barcelona, Spain.
| |
Collapse
|
5
|
Pant S, Cho BC, Kyriakopoulos CE, Spira A, Tannir N, Werner TL, Yan X, Neuteboom S, Chao R, Goel S. Targeting multiple receptor tyrosine kinases with sitravatinib: A Phase 1b study in advanced renal cell carcinoma and castrate-resistant prostate cancer. Invest New Drugs 2024:10.1007/s10637-024-01465-9. [PMID: 39168901 DOI: 10.1007/s10637-024-01465-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Sitravatinib (MGCD516) is an oral inhibitor of several closely related oncogenic tyrosine kinase receptors that include VEGFR-2 (vascular endothelial growth factor receptor-2), AXL, and MET (mesenchymal-epithelial transition). The safety and antitumor activity of sitravatinib are reported in patients from two histologic cohorts (anti-angiogenesis-refractory clear cell renal cell carcinoma [RCC] and castrate-resistant prostate cancer [CRPC] with bone metastases) who participated in a Phase 1/1b study. The patients were enrolled using a 3-stage design that was based on observed objective responses. Objective response rate (ORR) was the primary endpoint. Duration of response, progression-free survival (PFS), overall survival (OS), and safety were also assessed. Overall, 48 patients (RCC n = 38, CRPC n = 10) received ≥ 1 dose of sitravatinib. Both cohorts were heavily pretreated (median number of prior systemic therapies: RCC cohort 3, CRPC cohort 6). In the RCC cohort, ORR was 25.9%, P = 0.015 (null hypothesis [ORR ≤ 10%] was rejected). Responses were durable (median duration 13.2 months). Median PFS was 9.5 months and median OS was 30.0 months. No objective responses were seen in the CRPC cohort; median PFS and OS were 5.8 months and 10.1 months, respectively. Across both cohorts, diarrhea (72.9%), fatigue (54.2%), and hypertension (52.1%) were the most frequent all-cause treatment-emergent adverse events (TEAEs). Diarrhea and vomiting (both, 6.3%) were the most frequent serious TEAEs considered related to study treatment. Sitravatinib demonstrated an acceptable safety profile and promising clinical activity in patients with clear cell RCC refractory to prior angiogenesis inhibitor therapy. Strong indicators for clinical activity were not seen in patients with CRPC and bone metastases. Clinical trial registration:ClinicalTrials.gov NCT02219711.
Collapse
Affiliation(s)
- Shubham Pant
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | - Nizar Tannir
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Theresa L Werner
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | | | | | - Sanjay Goel
- Montefiore Medical Center, Bronx, NY, USA.
- Rutgers Cancer Institute, New Brunswick, NJ, USA.
| |
Collapse
|
6
|
Tanim K, Holtzhausen A, Thapa A, Huelse JM, Graham DK, Earp HS. MERTK Inhibition as a Targeted Novel Cancer Therapy. Int J Mol Sci 2024; 25:7660. [PMID: 39062902 PMCID: PMC11277220 DOI: 10.3390/ijms25147660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/06/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
In this issue honoring the contributions of Greg Lemke, the Earp and Graham lab teams discuss several threads in the discovery, action, signaling, and translational/clinical potential of MERTK, originally called c-mer, a member of the TYRO3, AXL, and MERTK (TAM) family of receptor tyrosine kinases. The 30-year history of the TAM RTK family began slowly as all three members were orphan RTKs without known ligands and/or functions when discovered by three distinct alternate molecular cloning strategies in the pre-genome sequencing era. The pace of understanding their physiologic and pathophysiologic roles has accelerated over the last decade. The activation of ligands bridging externalized phosphatidylserine (PtdSer) has placed these RTKs in a myriad of processes including neurodevelopment, cancer, and autoimmunity. The field is ripe for further advancement and this article hopefully sets the stage for further understanding and therapeutic intervention. Our review will focus on progress made through the collaborations of the Earp and Graham labs over the past 30 years.
Collapse
Affiliation(s)
- K.M. Tanim
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (K.M.T.); (A.T.); (J.M.H.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Alisha Holtzhausen
- Lineburger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Aashis Thapa
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (K.M.T.); (A.T.); (J.M.H.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Justus M. Huelse
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (K.M.T.); (A.T.); (J.M.H.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Douglas K. Graham
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (K.M.T.); (A.T.); (J.M.H.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - H. Shelton Earp
- Lineburger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
7
|
Demirsoy S, Tran H, Liu J, Li Y, Yang S, Aregawi D, Glantz MJ, Jacob NK, Walter V, Schell TD, Olmez I. Targeting Tyro3, Axl, and MerTK Receptor Tyrosine Kinases Significantly Sensitizes Triple-Negative Breast Cancer to CDK4/6 Inhibition. Cancers (Basel) 2024; 16:2253. [PMID: 38927958 PMCID: PMC11202171 DOI: 10.3390/cancers16122253] [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: 05/09/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype with high metastasis and mortality rates. Given the lack of actionable targets such as ER and HER2, TNBC still remains an unmet therapeutic challenge. Despite harboring high CDK4/6 expression levels, the efficacy of CDK4/6 inhibition in TNBC has been limited due to the emergence of resistance. The resistance to CDK4/6 inhibition is mainly mediated by RB1 inactivation. Since our aim is to overcome resistance to CDK4/6 inhibition, in this study, we primarily used the cell lines that do not express RB1. Following a screening for activated receptor tyrosine kinases (RTKs) upon CDK4/6 inhibition, we identified the TAM (Tyro3, Axl, and MerTK) RTKs as a crucial therapeutic vulnerability in TNBC. We show that targeting the TAM receptors with a novel inhibitor, sitravatinib, significantly sensitizes TNBC to CDK4/6 inhibitors. Upon prolonged HER2 inhibitor treatment, HER2+ breast cancers suppress HER2 expression, physiologically transforming into TNBC-like cells. We further show that the combined treatment is highly effective against drug-resistant HER2+ breast cancer as well. Following quantitative proteomics and RNA-seq data analysis, we extended our study into the immunophenotyping of TNBC. Given the roles of the TAM receptors in promoting the creation of an immunosuppressive tumor microenvironment (TME), we further demonstrate that the combination of CDK4/6 inhibitor abemaciclib and sitravatinib modifies the immune landscape of TNBC to favor immune checkpoint blockade. Overall, our study offers a novel and highly effective combination therapy against TNBC and potentially treatment-resistant HER2+ breast cancer that can be rapidly moved to the clinic.
Collapse
Affiliation(s)
- Seyma Demirsoy
- Departments of Neurosurgery, Penn State University, Hershey, PA 17033, USA (M.J.G.)
| | - Ha Tran
- Department of Radiation Oncology, Ohio State University, Columbus, OH 43210, USA
| | - Joseph Liu
- Department of Radiation Oncology, Ohio State University, Columbus, OH 43210, USA
| | - Yunzhan Li
- Departments of Cellular and Molecular Physiology, Penn State University, Hershey, PA 17033, USA
| | - Shengyu Yang
- Departments of Cellular and Molecular Physiology, Penn State University, Hershey, PA 17033, USA
| | - Dawit Aregawi
- Departments of Neurosurgery, Penn State University, Hershey, PA 17033, USA (M.J.G.)
| | - Michael J. Glantz
- Departments of Neurosurgery, Penn State University, Hershey, PA 17033, USA (M.J.G.)
| | | | - Vonn Walter
- Departments of Public Health Sciences, Penn State University, Hershey, PA 17033, USA
| | - Todd D. Schell
- Departments of Microbiology and Immunology, Penn State University, Hershey, PA 17033, USA
| | - Inan Olmez
- Departments of Neurosurgery, Penn State University, Hershey, PA 17033, USA (M.J.G.)
| |
Collapse
|
8
|
Dickinson A, Liu Y, Uvarov A, Peyret T, Marier JF, Chin C, Tran JQ. Concentration-QTc modeling of sitravatinib in patients with advanced solid malignancies. Clin Transl Sci 2024; 17:e13808. [PMID: 38700272 PMCID: PMC11067505 DOI: 10.1111/cts.13808] [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: 01/17/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 05/05/2024] Open
Abstract
Sitravatinib (MGCD516) is an orally available, small molecule, tyrosine kinase inhibitor that has been evaluated in patients with advanced solid tumors. Concentration-corrected QT interval (QTc; C-QTc) modeling was undertaken, using 767 matched concentration-ECG observations from 187 patients across two clinical studies in patients with advanced solid malignancies, across a dose range of 10-200 mg, via a linear mixed-effects (LME) model. The effect on heart rate (HR)-corrected QT interval via Fridericia's correction method (QTcF) at the steady-state maximum concentration (Cmax,ss) for the sitravatinib proposed therapeutic dosing regimen (100 mg malate once daily [q.d.]) without and with relevant intrinsic and extrinsic factors were predicted. No significant changes in HR from baseline were observed. Hysteresis between sitravatinib plasma concentration and change in QTcF from baseline (ΔQTcF) was not observed. There was no significant relationship between sitravatinib plasma concentration and ΔQTcF. The final C-QTc model predicted a mean (90% confidence interval [CI]) ΔQTcF of 3.92 (1.95-5.89) ms and 2.94 (0.23-6.10) ms at the proposed therapeutic dosing regimen in patients with normal organ function (best case scenario) and patients with hepatic impairment (worst-case scenario), respectively. The upper bounds of the 90% CIs were below the regulatory threshold of concern of 10 ms. The results of the described C-QTc analysis, along with corroborating results from nonclinical safety pharmacology studies, indicate that sitravatinib has a low risk of QTc interval prolongation at the proposed therapeutic dose of 100 mg malate q.d.
Collapse
Affiliation(s)
| | - Yong Liu
- Mirati Therapeutics IncSan DiegoCaliforniaUSA
| | - Artem Uvarov
- Certara Drug Development SolutionsPrincetonNew JerseyUSA
| | - Thomas Peyret
- Certara Drug Development SolutionsPrincetonNew JerseyUSA
| | - J. F. Marier
- Certara Drug Development SolutionsPrincetonNew JerseyUSA
| | - Curtis Chin
- Mirati Therapeutics IncSan DiegoCaliforniaUSA
| | | |
Collapse
|
9
|
Wang X, Pan H, Cui J, Chen X, Yoon WH, Carlino MS, Li X, Li H, Zhang J, Sun J, Guo J, Cui C. SAFFRON-103: a phase Ib study of sitravatinib plus tislelizumab in anti-PD-(L)1 refractory/resistant advanced melanoma. Immunotherapy 2024; 16:243-256. [PMID: 38197138 DOI: 10.2217/imt-2023-0130] [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] [Indexed: 01/11/2024] Open
Abstract
Aim: Investigate TKI sitravatinib plus anti-PD-1 antibody tislelizumab in patients with unresectable/advanced/metastatic melanoma with disease progression on/after prior first-line anti-PD-(L)1 monotherapy. Methods: Open-label, multicenter, multicohort study (NCT03666143). Patients in the melanoma cohort (N = 25) received sitravatinib once daily plus tislelizumab every 3 weeks. The primary end point was safety and tolerability. Results: Treatment-emergent adverse events (TEAEs) occurred in all patients, with ≥grade 3 TEAEs in 52.0%. Most TEAEs were mild-or-moderate in severity, none were fatal, and few patients discontinued treatment owing to TEAEs (12.0%). Objective response rate was 36.0% (95% CI: 18.0-57.5). Median progression-free survival was 6.7 months (95% CI: 4.1-not estimable). Conclusion: Sitravatinib plus tislelizumab had manageable safety/tolerability in patients with anti-PD-(L)1 refractory/resistant unresectable/advanced/metastatic melanoma, with promising antitumor activity. Clinical Trial Registration: NCT03666143 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Xuan Wang
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Hongming Pan
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiuwei Cui
- The First Hospital of Jilin University, Changchun, China
| | - Xiao Chen
- The First Hospital of Jilin University, Changchun, China
| | - Won-Hee Yoon
- Blacktown Cancer and Haematology Centre, Blacktown, NSW, Australia
| | - Matteo S Carlino
- Blacktown Cancer and Haematology Centre, Blacktown, NSW, Australia
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Xin Li
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Hui Li
- BeiGene (Shanghai) Co., Ltd., Shanghai, China
| | - Juan Zhang
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | | | - Jun Guo
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Chuanliang Cui
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
10
|
Zhao S, Ma Y, Liu L, Fang J, Ma H, Feng G, Xie B, Zeng S, Chang J, Ren J, Zhang Y, Xi N, Zhuang Y, Jiang Y, Zhang Q, Kang N, Zhang L, Zhao H. Ningetinib plus gefitinib in EGFR-mutant non-small-cell lung cancer with MET and AXL dysregulations: A phase 1b clinical trial and biomarker analysis. Lung Cancer 2024; 188:107468. [PMID: 38181454 DOI: 10.1016/j.lungcan.2024.107468] [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: 09/12/2023] [Revised: 12/26/2023] [Accepted: 01/01/2024] [Indexed: 01/07/2024]
Abstract
BACKGROUND MET and AXL dysregulations are implicated in acquired resistance to EGFR-TKIs in NSCLC. But consensus on the optimal definition for MET/AXL dysregulations in EGFR-mutant NSCLC is lacking. Here, we investigated the efficacy and tolerability of ningetinib (a MET/AXL inhibitor) plus gefitinib in EGFR-mutant NSCLC, and evaluated the clinical relevance of MET/AXL dysregulations by different definitions. METHODS Patients in this phase 1b dose-escalation/dose-expansion trial received ningetinib 30 mg/40 mg/60 mg plus gefitinib 250 mg once daily. Primary endpoints were tolerability (dose-escalation) and objective response rate (dose-expansion). MET/AXL status were analyzed using FISH and IHC. RESULTS Between March 2017 and January 2021, 108 patients were enrolled. The proportion of MET focal amplification, MET polysomy, MET overexpression, AXL amplification and AXL overexpression is 18.1 %, 5.6 %, 55.8 %, 8.1 % and 45.3 %, respectively. 6.8 % patients have concurrent MET amplification and AXL overexpression. ORR is 30.8 % for tumors with MET amplification, 0 % for MET polysomy, 24.1 % for MET overexpression, 20 % for AXL amplification and 27.6 % for AXL overexpression. For patients with concurrent MET amplification and AXL overexpression, ningetinib plus gefitinib provides an ORR of 80 %, DCR of 100 % and median PFS of 4.7 months. Tumors with higher MET copy number and AXL expression tend to have higher likelihood of response. Biomarker analyses show that MET focal amplification and overexpression are complementary in predicting clinical benefit from MET inhibition, while AXL dysregulations defined by an arbitrary level may dilute the efficacy of AXL blockade. CONCLUSIONS This study demonstrates that combined blockade of MET, AXL and EGFR is a feasible strategy for a subset of EGFR-mutant NSCLC. TRIAL REGISTRATION Chinadrugtrials.org.cn, CTR20160875.
Collapse
Affiliation(s)
- Shen Zhao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuxiang Ma
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lianke Liu
- Department of Oncology, Jiangsu Provincial Hospital, Nanjing, China
| | - Jian Fang
- Department of Thoracic Oncology, Beijing Cancer Hospital, Beijing, China
| | - Haiqing Ma
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Guosheng Feng
- Department of Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Bo Xie
- Department of Oncology, General Hospital of the PLA South Military Command, PLA, Guangzhou, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jianhua Chang
- Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jun Ren
- Department of Oncology, Beijing Shijitan Hospital, Beijing, China
| | | | - Ning Xi
- Sunshine Lake Pharma Co., Ltd, Dongguan, China; Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| | | | | | - Qi Zhang
- Sunshine Lake Pharma Co., Ltd, Dongguan, China
| | - Ning Kang
- Sunshine Lake Pharma Co., Ltd, Dongguan, China
| | - Li Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Hongyun Zhao
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
11
|
Borghaei H, de Marinis F, Dumoulin D, Reynolds C, Theelen WSME, Percent I, Gutierrez Calderon V, Johnson ML, Madroszyk-Flandin A, Garon EB, He K, Planchard D, Reck M, Popat S, Herbst RS, Leal TA, Shazer RL, Yan X, Harrigan R, Peters S. SAPPHIRE: phase III study of sitravatinib plus nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. Ann Oncol 2024; 35:66-76. [PMID: 37866811 DOI: 10.1016/j.annonc.2023.10.004] [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: 08/17/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Checkpoint inhibitor (CPI) therapy revolutionized treatment for advanced non-small-cell lung cancer (NSCLC); however, most patients progress due to primary or acquired resistance. Sitravatinib is a receptor tyrosine kinase inhibitor that can shift the immunosuppressive tumor microenvironment toward an immunostimulatory state. Combining sitravatinib with nivolumab (sitra + nivo) may potentially overcome initial CPI resistance. PATIENTS AND METHODS In the phase III SAPPHIRE study, patients with advanced non-oncogenic driven, nonsquamous NSCLC who initially benefited from (≥4 months on CPI without progression) and subsequently experienced disease progression on or after CPI combined with or following platinum-based chemotherapy were randomized 1 : 1 to sitra (100 mg once daily administered orally) + nivo (240 mg every 2 weeks or 480 mg every 4 weeks administered intravenously) or docetaxel (75 mg/m2 every 3 weeks administered intravenously). The primary endpoint was overall survival (OS). The secondary endpoints included progression-free survival (PFS), objective response rate (ORR), clinical benefit rate (CBR), duration of response (DOR; all assessed by blinded independent central review), and safety. RESULTS A total of 577 patients included randomized: sitra + nivo, n = 284; docetaxel, n = 293 (median follow-up, 17.1 months). Sitra + nivo did not significantly improve OS versus docetaxel [median, 12.2 versus 10.6 months; hazard ratio (HR) 0.86, 95% confidence interval (CI) 0.70-1.05; P = 0.144]. The median PFS was 4.4 versus 5.4 months, respectively (HR 1.08, 95% CI 0.89-1.32; P = 0.452). The ORR was 15.6% for sitra + nivo and 17.2% for docetaxel (P = 0.597); CBR was 75.5% and 64.5%, respectively (P = 0.004); median DOR was 7.4 versus 7.1 months, respectively (P = 0.924). Grade ≥3 treatment-related adverse events were observed in 53.0% versus 66.7% of patients receiving sitra + nivo versus docetaxel, respectively. CONCLUSIONS Although median OS was numerically longer with sitra + nivo, the primary endpoint was not met in patients with previously treated advanced nonsquamous NSCLC. The safety profiles demonstrated were consistent with previous reports.
Collapse
Affiliation(s)
- H Borghaei
- Hematology and Oncology Department, Fox Chase Cancer Center, Philadelphia, USA.
| | - F de Marinis
- Division of Thoracic Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - D Dumoulin
- Department of Pulmonary Medicine, Erasmus Medisch Centrum, Rotterdam, the Netherlands
| | - C Reynolds
- Ocala Cancer Center, Florida Cancer Specialists and Research Institute - North Region (SCRI), Ocala, USA
| | - W S M E Theelen
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - I Percent
- North Port Cancer Center, Florida Cancer Specialists and Research Institute - South Region (SCRI), Port Charlotte, USA
| | - V Gutierrez Calderon
- Department of Medical Oncology, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - M L Johnson
- Department of Medical Oncology, Sarah Cannon Research Institute, Tennessee Oncology, Nashville, USA
| | | | - E B Garon
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles
| | - K He
- Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, USA
| | - D Planchard
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France
| | - M Reck
- Department of Thoracic Oncology, LungenClinic, Airway Research Center North, German Center for Lung Research, Grosshansdorf, Germany
| | - S Popat
- Lung Unit, Department of Medicine, The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London, UK
| | - R S Herbst
- Section of Medical Oncology, Yale University, New Haven
| | - T A Leal
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta
| | - R L Shazer
- Department of Clinical Research and Development, Mirati Therapeutics, Inc., San Diego, USA
| | - X Yan
- Department of Clinical Research and Development, Mirati Therapeutics, Inc., San Diego, USA
| | - R Harrigan
- Department of Clinical Research and Development, Mirati Therapeutics, Inc., San Diego, USA
| | - S Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| |
Collapse
|
12
|
Thet D, Areepium N, Siritientong T. Effects of Probiotics on Chemotherapy-induced Diarrhea. Nutr Cancer 2023; 75:1811-1821. [PMID: 37908158 DOI: 10.1080/01635581.2023.2267779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 11/02/2023]
Abstract
Chemotherapy-induced diarrhea (CID) is a common adverse event in cancer patients, which, unless treated, may lead to drug discontinuation and treatment failure. Some probiotics such as Lactobacillus, Bifidobacterium, and Saccharomyces species have been gaining clinical attention in alleviating chemotherapy-induced adverse events including diarrhea. This comprehensive review provides an overview and discusses preventive approaches of probiotics with respect to CID in several types of cancers. The potential mechanisms of probiotics may comprise regulation of intestinal microbiota, modulation of immune functions, or reduction of proinflammatory cytokines. The efficacy and safety precautions of probiotics in immunocompromised cancer patients are discussed. The non-pharmacological strategy using probiotics could reduce the use of anti-diarrheal or antibiotic agents. Some individuals experienced shorter length of hospital stay, better gastrointestinal function, and reduced incidence of chemotherapy dose reduction after probiotic administration. Nonetheless, some studies failed to report the benefits of probiotics in certain patients. This review also highlights preventive protocols and therapeutic implications by considering the potential influencing factors, particularly types of probiotic strains, dosages of probiotics, duration of their administration, patients' tolerability, and variations in probiotic effects over the cancer stages.
Collapse
Affiliation(s)
- Daylia Thet
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Nutthada Areepium
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tippawan Siritientong
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Metabolomics for Life Sciences Research Unit, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
13
|
Ubil E, Zahid KR. Structure and functions of Mer, an innate immune checkpoint. Front Immunol 2023; 14:1244170. [PMID: 37936688 PMCID: PMC10626544 DOI: 10.3389/fimmu.2023.1244170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Immunotherapy is a promising therapeutic tool that promotes the elimination of cancerous cells by a patient's own immune system. However, in the clinical setting, the number of cancer patients benefitting from immunotherapy is limited. Identification and targeting of other immune subsets, such as tumor-associated macrophages, and alternative immune checkpoints, like Mer, may further limit tumor progression and therapy resistance. In this review, we highlight the key roles of macrophage Mer signaling in immune suppression. We also summarize the role of pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes in tumor onset and progression and how Mer structure and activation can be targeted therapeutically to alter activation state. Preclinical and clinical studies focusing on Mer kinase inhibition have demonstrated the potential of targeting this innate immune checkpoint, leading to improved anti-tumor responses and patient outcomes.
Collapse
Affiliation(s)
- Eric Ubil
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | | |
Collapse
|
14
|
Nguyen TM, Ngoc DTM, Choi JH, Lee CH. Unveiling the Neural Environment in Cancer: Exploring the Role of Neural Circuit Players and Potential Therapeutic Strategies. Cells 2023; 12:1996. [PMID: 37566075 PMCID: PMC10417274 DOI: 10.3390/cells12151996] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
The regulation of the immune environment within the tumor microenvironment has provided new opportunities for cancer treatment. However, an important microenvironment surrounding cancer that is often overlooked despite its significance in cancer progression is the neural environment surrounding the tumor. The release of neurotrophic factors from cancer cells is implicated in cancer growth and metastasis by facilitating the infiltration of nerve cells into the tumor microenvironment. This nerve-tumor interplay can elicit cancer cell proliferation, migration, and invasion in response to neurotransmitters. Moreover, it is possible that cancer cells could establish a network resembling that of neurons, allowing them to communicate with one another through neurotransmitters. The expression levels of players in the neural circuits of cancers could serve as potential biomarkers for cancer aggressiveness. Notably, the upregulation of certain players in the neural circuit has been linked to poor prognosis in specific cancer types such as breast cancer, pancreatic cancer, basal cell carcinoma, and stomach cancer. Targeting these players with inhibitors holds great potential for reducing the morbidity and mortality of these carcinomas. However, the efficacy of anti-neurogenic agents in cancer therapy remains underexplored, and further research is necessary to evaluate their effectiveness as a novel approach for cancer treatment. This review summarizes the current knowledge on the role of players in the neural circuits of cancers and the potential of anti-neurogenic agents for cancer therapy.
Collapse
Affiliation(s)
- Tuan Minh Nguyen
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| | - Dinh Thi Minh Ngoc
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| | - Jung-Hye Choi
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Chang-Hoon Lee
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| |
Collapse
|
15
|
Okoye C, Tran M, Soladoye E, Akahara DE, Emeasoba CM, Ojinna BT, Anasonye E, Obadare OO, Diala CS, Salaudeen BH, Evbayekha EO, Okobi OE. A Review of 10-Year Survivability of Immunotherapy in the Management of Colon Cancer. Cureus 2023; 15:e43189. [PMID: 37692610 PMCID: PMC10485874 DOI: 10.7759/cureus.43189] [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: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Colon cancer is one of the most common cancers in the United States of America. In addition to conventional treatment approaches such as surgery, chemotherapy, and radiation for colorectal cancer, immunotherapy has gained recognition over the past few years. However, its effectiveness in colorectal cancer treatment is controversial. Our study investigates the survival and progression-free rates of immunotherapy for different types of colorectal cancer over the last 10 years. We conducted literature reviews from various clinical trials and research studies to evaluate immunotherapy's role in colorectal cancer treatment. We also investigated how it affects clinical outcomes. We discovered a range of effective immunotherapy approaches targeting various growth factors and signaling pathways. These modalities include monoclonal antibodies aimed at growth factors such as epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), human epidermal growth factor receptor 2 (HER2), and downstream signaling pathways such as mitogen-activated protein kinase (MAPK), kirsten rat sarcoma viral oncogene (KRAS), B-raf proto-oncogene, serine/threonine kinase (BRAF), and phosphatase and tensin homolog (PTEN). Additionally, we identified immune checkpoint inhibitors, such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) inhibitors and programmed cell death ligand 1 (PD-L1) inhibitors, as well as target therapy and adoptive cell therapy as promising immunotherapeutic options. Nevertheless, the application of immunotherapy remains highly limited due to various factors influencing survival and progression-free rates, including tumor microenvironment, microsatellite instability, immune checkpoint expression, and gut microbiome. Additionally, its effectiveness is restricted to a small subgroup of patients, accompanied by side effects and the development of drug resistance mechanisms. To unlock its full potential, further clinical trials and research on molecular pathways in colorectal cancer are imperative. This will ultimately enhance drug discovery success and lead to more effective clinical management approaches.
Collapse
Affiliation(s)
- Chiugo Okoye
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - My Tran
- Internal Medicine, Baptist Health-University of Arkansas for Medical Sciences - Arkansas, North Little Rock, USA
| | | | | | | | - Blessing T Ojinna
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | | | | - Chiamaka S Diala
- Health/Biomedical Informatics, The University of Texas Health Science Center, Houston, USA
- Internal Medicine, Piedmont Athens Regional, Athens, USA
| | | | | | - Okelue E Okobi
- Family Medicine, Medficient Health Systems, Laurel, USA
- Family Medicine, Lakeside Medical Center, Belle Glade, USA
| |
Collapse
|
16
|
Minchom A, Popat S. Sitravatinib and Acquired Immune Checkpoint Inhibitor Resistance: A Gem for the Future? J Thorac Oncol 2023; 18:830-833. [PMID: 37348988 DOI: 10.1016/j.jtho.2023.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 06/24/2023]
Affiliation(s)
- Anna Minchom
- Section of Clinical Trials, Institute of Cancer Research, London, United Kingdom; Lung Unit, Royal Marsden Hospital, London, United Kingdom
| | - Sanjay Popat
- Section of Clinical Trials, Institute of Cancer Research, London, United Kingdom; Lung Unit, Royal Marsden Hospital, London, United Kingdom.
| |
Collapse
|
17
|
He K, Berz D, Gadgeel SM, Iams WT, Bruno DS, Blakely CM, Spira AI, Patel MR, Waterhouse DM, Richards DA, Pham A, Jotte R, Hong DS, Garon EB, Traynor A, Olson P, Latven L, Yan X, Shazer R, Leal TA. MRTX-500 Phase 2 Trial: Sitravatinib With Nivolumab in Patients With Nonsquamous NSCLC Progressing On or After Checkpoint Inhibitor Therapy or Chemotherapy. J Thorac Oncol 2023; 18:907-921. [PMID: 36842467 PMCID: PMC10330304 DOI: 10.1016/j.jtho.2023.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
INTRODUCTION Sitravatinib, a receptor tyrosine kinase inhibitor targeting TYRO3, AXL, MERTK receptors, and vascular epithelial growth factor receptor 2, can shift the tumor microenvironment toward an immunostimulatory state. Combining sitravatinib with checkpoint inhibitors (CPIs) may augment antitumor activity. METHODS The phase 2 MRTX-500 study evaluated sitravatinib (120 mg daily) with nivolumab (every 2 or 4 wk) in patients with advanced nonsquamous NSCLC who progressed on or after previous CPI (CPI-experienced) or chemotherapy (CPI-naive). CPI-experienced patients had a previous clinical benefit (PCB) (complete response, partial response, or stable disease for at least 12 weeks then disease progression) or no PCB (NPCB) from CPI. The primary end point was objective response rate (ORR); secondary objectives included safety and secondary efficacy end points. RESULTS Overall, 124 CPI-experienced (NPCB, n = 35; PCB, n = 89) and 32 CPI-naive patients were treated. Investigator-assessed ORR was 11.4% in patients with NPCB, 16.9% with PCB, and 25.0% in CPI-naive. The median progression-free survival was 3.7, 5.6, and 7.1 months with NPCB, PCB, and CPI-naive, respectively; the median overall survival was 7.9 and 13.6 months with NPCB and PCB, respectively (not reached in CPI-naive patients; median follow-up 20.4 mo). Overall, (N = 156), any grade treatment-related adverse events (TRAEs) occurred in 93.6%; grade 3/4 in 58.3%. One grade 5 TRAE occurred in a CPI-naive patient. TRAEs led to treatment discontinuation in 14.1% and dose reduction or interruption in 42.9%. Biomarker analyses supported an immunostimulatory mechanism of action. CONCLUSIONS Sitravatinib with nivolumab had a manageable safety profile. Although ORR was not met, this combination exhibited antitumor activity and encouraged survival in CPI-experienced patients with nonsquamous NSCLC.
Collapse
Affiliation(s)
- Kai He
- Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, Ohio.
| | - David Berz
- Department of Cellular Therapeutics, Beverly Hills Cancer Center, Beverly Hills, California; Current Affiliation: Valkyrie Clinical Trials, Los Angeles, California
| | - Shirish M Gadgeel
- Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan
| | - Wade T Iams
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee
| | - Debora S Bruno
- University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Collin M Blakely
- Department of Medicine, University of California San Francisco, San Francisco, California; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Alexander I Spira
- Virginia Cancer Specialists, Fairfax, Virginia; US Oncology Network, The Woodlands, Texas
| | - Manish R Patel
- Division Of Hematology, Oncology and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| | - David M Waterhouse
- US Oncology Network, The Woodlands, Texas; Department of Clinical Research, Oncology Hematology Care, Cincinnati, Ohio; Current affiliation: Dana-Farber/Brigham and Women's Cancer Center at Milford Regional Medical Center, Milford, Massachusetts
| | - Donald A Richards
- US Oncology Network, The Woodlands, Texas; Texas Oncology, Tyler, Texas
| | | | - Robert Jotte
- US Oncology Network, The Woodlands, Texas; Rocky Mountain Cancer Centers, Denver, Colorado
| | - David S Hong
- MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Edward B Garon
- Department Of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Anne Traynor
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Peter Olson
- Mirati Therapeutics, Inc., San Diego, California
| | - Lisa Latven
- Mirati Therapeutics, Inc., San Diego, California
| | - Xiaohong Yan
- Mirati Therapeutics, Inc., San Diego, California
| | | | - Ticiana A Leal
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin; Current Affiliation: Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
18
|
Ye W, Li M, Luo K. Therapies Targeting Immune Cells in Tumor Microenvironment for Non-Small Cell Lung Cancer. Pharmaceutics 2023; 15:1788. [PMID: 37513975 PMCID: PMC10384189 DOI: 10.3390/pharmaceutics15071788] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
The tumor microenvironment (TME) plays critical roles in immune modulation and tumor malignancies in the process of cancer development. Immune cells constitute a significant component of the TME and influence the migration and metastasis of tumor cells. Recently, a number of therapeutic approaches targeting immune cells have proven promising and have already been used to treat different types of cancer. In particular, PD-1 and PD-L1 inhibitors have been used in the first-line setting in non-small cell lung cancer (NSCLC) with PD-L1 expression ≥1%, as approved by the FDA. In this review, we provide an introduction to the immune cells in the TME and their efficacies, and then we discuss current immunotherapies in NSCLC and scientific research progress in this field.
Collapse
Affiliation(s)
- Wei Ye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
| | - Meiye Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
| | - Kewang Luo
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
- People's Hospital of Longhua, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen 518109, China
| |
Collapse
|
19
|
Jang A, Lanka SM, Huang M, Casado CV, Caputo SA, Sweeney PL, Gupta K, Pocha O, Habibian N, Hawkins ME, Lieberman AD, Schwartz J, Jaeger EB, Miller PJ, Layton JL, Barata PC, Lewis BE, Ledet EM, Sartor O. Comparison of circulating tumor DNA between African American and Caucasian patients with metastatic castrate-resistant prostate cancer post-abiraterone and/or enzalutamide. Prostate 2023. [PMID: 37113064 DOI: 10.1002/pros.24544] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/18/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND African American men are much more likely than Caucasian men to be diagnosed with and to die of prostate cancer. Genetic differences likely play a role. The cBioPortal database reveals that African American men with prostate cancer have higher rates of CDK12 somatic mutations compared to Caucasian men. However, this does not account for prior prostate cancer treatments, which are particularly important in the castrate-resistant setting. We aimed to compare somatic mutations based on circulating tumor DNA (ctDNA) in metastatic castration-resistant prostate cancer (mCRPC) between African American and Caucasian men after exposure to abiraterone and/or enzalutamide. METHODS This single-institution retrospective study characterizes the somatic mutations detected on ctDNA for African American and Caucasian men with mCRPC who had progressed after abiraterone and/or enzalutamide from 2015 through 2022. We evaluated the gene mutations and types of mutations in this mCRPC cohort. RESULTS There were 50 African American and 200 Caucasian men with CRPC with available ctDNA data. African American men were younger at the time of diagnosis (p = 0.008) and development of castration resistance (p = 0.006). African American men were more likely than Caucasian men to have pathogenic/likely pathogenic (P/LP) mutations in CDK12 (12% vs. 1.5%; p = 0.003) and copy number amplifications and P/LP mutations in KIT (8.0% vs. 1.5%; p = 0.031). African American men were also significantly more likely to have frameshift mutations (28% vs. 14%; p = 0.035). CONCLUSIONS Compared to Caucasian men, African American men with mCRPC after exposure to abiraterone and/or enzalutamide had a higher incidence of somatic CDK12 P/LP mutations and KIT amplifications and P/LP mutations based on ctDNA. African American men also had more frameshift mutations. We hypothesize that these findings have potential implications for tumor immunogenicity.
Collapse
Affiliation(s)
- Albert Jang
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Sree M Lanka
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Minqi Huang
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Crystal V Casado
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Sydney A Caputo
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Patrick L Sweeney
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Kanika Gupta
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Olivia Pocha
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | | - Madeline E Hawkins
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Alexandra D Lieberman
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jennifer Schwartz
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Ellen B Jaeger
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Patrick J Miller
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jodi L Layton
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Pedro C Barata
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Brian E Lewis
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Elisa M Ledet
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Oliver Sartor
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
20
|
Gong H, Xu HM, Zhang DK. Focusing on discoidin domain receptors in premalignant and malignant liver diseases. Front Oncol 2023; 13:1123638. [PMID: 37007062 PMCID: PMC10050580 DOI: 10.3389/fonc.2023.1123638] [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: 12/14/2022] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
Discoidin domain receptors (DDRs) are receptor tyrosine kinases on the membrane surface that bind to extracellular collagens, but they are rarely expressed in normal liver tissues. Recent studies have demonstrated that DDRs participate in and influence the processes underlying premalignant and malignant liver diseases. A brief overview of the potential roles of DDR1 and DDR2 in premalignant and malignant liver diseases is presented. DDR1 has proinflammatory and profibrotic benefits and promotes the invasion, migration and liver metastasis of tumour cells. However, DDR2 may play a pathogenic role in early-stage liver injury (prefibrotic stage) and a different role in chronic liver fibrosis and in metastatic liver cancer. These views are critically significant and first described in detail in this review. The main purpose of this review was to describe how DDRs act in premalignant and malignant liver diseases and their potential mechanisms through an in-depth summary of preclinical in vitro and in vivo studies. Our work aims to provide new ideas for cancer treatment and accelerate translation from bench to bedside.
Collapse
Affiliation(s)
| | | | - De-Kui Zhang
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
21
|
Zhao J, Yu X, Huang D, Ma Z, Gao B, Cui J, Chu Q, Zhou Q, Sun M, Day D, Wu J, Pan H, Wang L, Voskoboynik M, Wang Z, Liu Y, Li H, Zhang J, Peng Y, Wu YL. SAFFRON-103: a phase 1b study of the safety and efficacy of sitravatinib combined with tislelizumab in patients with locally advanced or metastatic non-small cell lung cancer. J Immunother Cancer 2023; 11:jitc-2022-006055. [PMID: 36808075 PMCID: PMC9944269 DOI: 10.1136/jitc-2022-006055] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Some patients with locally advanced/metastatic non-small cell lung cancer (NSCLC) respond poorly to anti-programmed cell death protein 1 (PD-1)/anti-programmed death-ligand 1 (PD-L1) treatments. Combination with other agents may improve the outcomes. This open-label, multicenter, phase 1b trial investigated the combination of sitravatinib, a spectrum-selective tyrosine kinase inhibitor, plus anti-PD-1 antibody tislelizumab. METHODS Patients with locally advanced/metastatic NSCLC were enrolled (Cohorts A, B, F, H, and I; N=22-24 per cohort). Cohorts A and F included patients previously treated with systemic therapy, with anti-PD-(L)1-resistant/refractory non-squamous (cohort A) or squamous (cohort F) disease. Cohort B included patients previously treated with systemic therapy, with anti-PD-(L)1-naïve non-squamous disease. Cohorts H and I included patients without prior systemic therapy for metastatic disease, no prior anti-PD-(L)1/immunotherapy, with PD-L1-positive non-squamous (cohort H) or squamous (cohort I) histology. Patients received sitravatinib 120 mg orally one time per day plus tislelizumab 200 mg intravenously every 3 weeks, until study withdrawal, disease progression, unacceptable toxicity, or death. The primary endpoint was safety/tolerability among all treated patients (N=122). Secondary endpoints included investigator-assessed tumor responses and progression-free survival (PFS). RESULTS Median follow-up was 10.9 months (range: 0.4-30.6). Treatment-related adverse events (TRAEs) occurred in 98.4% of the patients, with ≥Grade 3 TRAEs in 51.6%. TRAEs led to discontinuation of either drug in 23.0% of the patients. Overall response rate was 8.7% (n/N: 2/23; 95% CI: 1.1% to 28.0%), 18.2% (4/22; 95% CI: 5.2% to 40.3%), 23.8% (5/21; 95% CI: 8.2% to 47.2%), 57.1% (12/21; 95% CI: 34.0% to 78.2%), and 30.4% (7/23; 95% CI: 13.2% to 52.9%) in cohorts A, F, B, H, and I, respectively. Median duration of response was not reached in cohort A and ranged from 6.9 to 17.9 months across other cohorts. Disease control was achieved in 78.3-90.9% of the patients. Median PFS ranged from 4.2 (cohort A) to 11.1 months (cohort H). CONCLUSIONS In patients with locally advanced/metastatic NSCLC, sitravatinib plus tislelizumab was tolerable for most patients, with no new safety signals and overall safety profiles consistent with known profiles of these agents. Objective responses were observed in all cohorts, including in patients naïve to systemic and anti-PD-(L)1 treatments, or with anti-PD-(L)1 resistant/refractory disease. Results support further investigation in selected NSCLC populations. TRIAL REGISTRATION NUMBER NCT03666143.
Collapse
Affiliation(s)
- Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xinmin Yu
- Department of Medical Oncology, Cancer Hospital of University of Chinese Academy of Sciences & Zhejiang Cancer Hospital, Hangzhou, China
| | - Dingzhi Huang
- Department of Thoracic Medical Oncology, Tianjin Cancer Hospital, Tianjin, China
| | - Zhiyong Ma
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University; Henan Cancer Hospital, Zhengzhou, China
| | - Bo Gao
- Blacktown Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Wuhan, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Meili Sun
- Department of Oncology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Daphne Day
- Medical Oncology, Monash Health and Monash University, Melbourne, Victoria, Australia
| | - Jingxun Wu
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | | | - Mark Voskoboynik
- Medical Oncology, Nucleus Network, Melbourne, VIC, Australia and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Zhehai Wang
- Department of Internal Medicine - Oncology, Shandong Cancer Hospital & Institute, Jinan, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Hui Li
- BeiGene (Shanghai) Co., Ltd, Shanghai, China
| | - Juan Zhang
- BeiGene (Beijing) Co., Ltd, Beijing, China
| | - Yanyan Peng
- BeiGene (Shanghai) Co., Ltd, Shanghai, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
22
|
Zhang Y, Wang P, Wang Y, Shen Y. Sitravatinib as a potent FLT3 inhibitor can overcome gilteritinib resistance in acute myeloid leukemia. Biomark Res 2023; 11:8. [PMID: 36691065 PMCID: PMC9872318 DOI: 10.1186/s40364-022-00447-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/29/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Gilteritinib is the only drug approved as monotherapy for acute myeloid leukemia (AML) patients harboring FMS-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) mutation throughout the world. However, drug resistance inevitably develops in clinical. Sitravatinib is a multi-kinase inhibitor under evaluation in clinical trials of various solid tumors. In this study, we explored the antitumor activity of sitravatinib against FLT3-ITD and clinically-relevant drug resistance in FLT3 mutant AML. METHODS Growth inhibitory assays were performed in AML cell lines and BaF3 cells expressing various FLT3 mutants to evaluate the antitumor activity of sitravatinib in vitro. Immunoblotting was used to examine the activity of FLT3 and its downstream pathways. Molecular docking was performed to predict the binding sites of FLT3 to sitravatinib. The survival benefit of sitravatinib in vivo was assessed in MOLM13 xenograft mouse models and mouse models of transformed BaF3 cells harboring different FLT3 mutants. Primary patient samples and a patient-derived xenograft (PDX) model were also used to determine the efficacy of sitravatinib. RESULTS Sitravatinib inhibited cell proliferation, induced cell cycle arrest and apoptosis in FLT3-ITD AML cell lines. In vivo studies showed that sitravatinib exhibited a better therapeutic effect than gilteritinib in MOLM13 xenograft model and BaF3-FLT3-ITD model. Unlike gilteritinib, the predicted binding sites of sitravatinib to FLT3 did not include F691 residue. Sitravatinib displayed a potent inhibitory effect on FLT3-ITD-F691L mutation which conferred resistance to gilteritinib and all other FLT3 inhibitors available, both in vitro and in vivo. Compared with gilteritinib, sitravatinib retained effective activity against FLT3 mutation in the presence of cytokines through the more potent and steady inhibition of p-ERK and p-AKT. Furthermore, patient blasts harboring FLT3-ITD were more sensitive to sitravatinib than to gilteritinib in vitro and in the PDX model. CONCLUSIONS Our study reveals the potential therapeutic role of sitravatinib in FLT3 mutant AML and provides an alternative inhibitor for the treatment of AML patients who are resistant to current FLT3 inhibitors.
Collapse
Affiliation(s)
- Yvyin Zhang
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Peihong Wang
- Department of Hematology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000 China
| | - Yang Wang
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Yang Shen
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
23
|
Prasad Shenoy G, Pal R, Gurubasavaraja Swamy P, Singh E, Manjunathaiah Raghavendra N, Sanjay Dhiwar P. Discoidin Domain Receptor Inhibitors as Anticancer Agents: A Systematic Review on Recent Development of DDRs Inhibitors, their Resistance and Structure Activity Relationship. Bioorg Chem 2022; 130:106215. [DOI: 10.1016/j.bioorg.2022.106215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Accepted: 10/15/2022] [Indexed: 11/02/2022]
|
24
|
The Role of Neural Signaling in the Pancreatic Cancer Microenvironment. Cancers (Basel) 2022; 14:cancers14174269. [PMID: 36077804 PMCID: PMC9454556 DOI: 10.3390/cancers14174269] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Pancreatic cancer is a highly lethal malignant disease with a dense stroma, called the tumor microenvironment. Accumulating evidence indicates the important role of sympathetic, parasympathetic, and sensory nerves in the tumor microenvironment of various cancers, including pancreatic cancer. Cancer cells and neural cells interact with each other to form a complex network and cooperatively promote cancer growth and invasion. In this review article, we describe the current understanding of the role of nerves in the tumor microenvironment. Abstract Pancreatic cancer is one of the most lethal malignant diseases. Various cells in the tumor microenvironment interact with tumor cells and orchestrate to support tumor progression. Several kinds of nerves are found in the tumor microenvironment, and each plays an essential role in tumor biology. Recent studies have shown that sympathetic, parasympathetic, and sensory neurons are found in the pancreatic cancer microenvironment. Neural signaling not only targets neural cells, but tumor cells and immune cells via neural receptors expressed on these cells, through which tumor growth, inflammation, and anti-tumor immunity are affected. Thus, these broad-range effects of neural signaling in the pancreatic cancer microenvironment may represent novel therapeutic targets. The modulation of neural signaling may be a therapeutic strategy targeting the whole tumor microenvironment. In this review, we describe the current understanding of the role of nerves in the tumor microenvironment of various cancers, with an emphasis on pancreatic cancer. We also discuss the underlying mechanisms and the possibility of therapeutic applications.
Collapse
|