1
|
Qiu C, Sun N, Zeng S, Chen L, Gong F, Tian J, Xiong Y, Peng L, He H, Ming Y. Unveiling the therapeutic promise of EphA2 in glioblastoma: a comprehensive review. Discov Oncol 2024; 15:501. [PMID: 39331302 PMCID: PMC11436538 DOI: 10.1007/s12672-024-01380-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Glioblastoma (GBM), a primary brain tumor, exhibits remarkable invasiveness and is characterized by its intricate location, infiltrative behavior, the presence of both the blood-brain barrier (BBB) and the blood-brain tumor barrier (BBTB), phenotypic diversity, an immunosuppressive microenvironment with limited development yet rich vascularity, as well as the resistant nature of glioblastoma stem cells (GSCs) towards traditional chemotherapy and radiotherapy. These formidable factors present substantial obstacles in the quest for effective GBM treatments. Following extensive research spanning three decades, the hepatocellular receptor A2 (EphA2) receptor tyrosine kinase has emerged as a promising molecular target with translational potential in the realm of cancer therapy. Numerous compounds aimed at targeting EphA2 have undergone rigorous evaluation and clinical investigation. This article provides a comprehensive account of the distinctive roles played by canonical and non-canonical EphA2 signaling in various contexts, while also exploring the involvement of the EphA2-ephrin A1 signaling axis in GBM pathogenesis. Additionally, the review offers an overview of completed clinical trials targeting EphA2 for GBM treatment, shedding light on both the prospects and challenges associated with EphA2-directed interventions in the domain of cancer therapeutics.
Collapse
Affiliation(s)
- Caohang Qiu
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Ning Sun
- Department of Pediatric Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shan Zeng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Feilong Gong
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Junjie Tian
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Yu Xiong
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Lilei Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Haiping He
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Yang Ming
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
- Sichuan Clinical Research Center of Neurosurgery, Luzhou, 646000, People's Republic of China.
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, People's Republic of China.
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| |
Collapse
|
2
|
Pflug KM, Lee DW, Tripathi A, Bankaitis VA, Burgess K, Sitcheran R. Cyanine Dye Conjugation Enhances Crizotinib Localization to Intracranial Tumors, Attenuating NF-κB-Inducing Kinase Activity and Glioma Progression. Mol Pharm 2023; 20:6140-6150. [PMID: 37939020 DOI: 10.1021/acs.molpharmaceut.3c00496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Glioblastoma (GBM) is a highly aggressive form of brain cancer with a poor prognosis and limited treatment options. The ALK and c-MET inhibitor Crizotinib has demonstrated preclinical therapeutic potential for newly diagnosed GBM, although its efficacy is limited by poor penetration of the blood brain barrier. Here, we identify Crizotinib as a novel inhibitor of nuclear factor-κB (NF-κB)-inducing kinase, which is a key regulator of GBM growth and proliferation. We further show that the conjugation of Crizotinib to a heptamethine cyanine dye, or a near-infrared dye (IR-Crizotinib), attenuated glioma cell proliferation and survival in vitro to a greater extent than unconjugated Crizotinib. Moreover, we observed increased IR-Crizotinib localization to orthotopic mouse xenograft GBM tumors, which resulted in impaired tumor growth in vivo. Overall, IR-Crizotinib exhibited improved intracranial chemotherapeutic delivery and tumor localization with concurrent inhibition of NIK and noncanonical NF-κB signaling, thereby reducing glioma growth in vitro, as well as in vivo, and increasing survival in a preclinical rodent model.
Collapse
Affiliation(s)
- Kathryn M Pflug
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Dong W Lee
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Ashutosh Tripathi
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Vytas A Bankaitis
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Kevin Burgess
- Department of Chemistry, Texas A&M University, Box 30012, College Station, Texas 77842, United States
| | - Raquel Sitcheran
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| |
Collapse
|
3
|
Tyrosine Kinase Inhibitors for Glioblastoma Multiforme: Challenges and Opportunities for Drug Delivery. Pharmaceutics 2022; 15:pharmaceutics15010059. [PMID: 36678688 PMCID: PMC9863099 DOI: 10.3390/pharmaceutics15010059] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain tumor with high mortality rates. Due to its invasiveness, heterogeneity, and incomplete resection, the treatment is very challenging. Targeted therapies such as tyrosine kinase inhibitors (TKIs) have great potential for GBM treatment, however, their efficacy is primarily limited by poor brain distribution due to the presence of the blood-brain barrier (BBB). This review focuses on the potential of TKIs in GBM therapy and provides an insight into the reasons behind unsuccessful clinical trials of TKIs in GBM despite the success in treating other cancer types. The main section is dedicated to the use of promising drug delivery strategies for targeted delivery to brain tumors. Use of brain targeted delivery strategies can help enhance the efficacy of TKIs in GBM. Among various drug delivery approaches used to bypass or cross BBB, utilizing nanocarriers is a promising strategy to augment the pharmacokinetic properties of TKIs and overcome their limitations. This is because of their advantages such as the ability to cross BBB, chemical stabilization of drug in circulation, passive or active targeting of tumor, modulation of drug release from the carrier, and the possibility to be delivered via non-invasive intranasal route.
Collapse
|
4
|
Wang R, Zhao L, Wang S, Zhao X, Liang C, Wang P, Li D. Regulatory pattern of abnormal promoter CpG island methylation in the glioblastoma multiforme classification. Front Genet 2022; 13:989985. [PMID: 36199581 PMCID: PMC9527345 DOI: 10.3389/fgene.2022.989985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/30/2022] [Indexed: 11/15/2022] Open
Abstract
Glioblastoma (GBM) is characterized by extensive genetic and phenotypic heterogeneity. However, it remains unexplored primarily how CpG island methylation abnormalities in promoter mediate glioblastoma typing. First, we presented a multi-omics scale map between glioblastoma sample clusters constructed based on promoter CpG island (PCGI) methylation-driven genes, using datasets including methylation profiles, expression profiles, and single-cell sequencing data from multiple highly annotated public clinical cohorts. Second, we identified differences in the tumor microenvironment between the two glioblastoma sample clusters and resolved key signaling pathways between cell clusters at the single-cell level based on comprehensive comparative analyses to investigate the reasons for survival differences between two of these clusters. Finally, we developed a diagnostic map and a prediction model for glioblastoma, and compared theoretical differences of drug sensitivity between two glioblastoma sample clusters. In summary, this study established a classification system for dissecting promoter CpG island methylation heterogeneity in glioblastoma and provides a new perspective for the diagnosis and treatment of glioblastoma.
Collapse
Affiliation(s)
- Rendong Wang
- School of Biomedical Engineering, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing, China
| | - Lei Zhao
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shijia Wang
- School of Biomedical Engineering, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing, China
| | - Xiaoxiao Zhao
- School of Biomedical Engineering, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing, China
| | - Chuanyu Liang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Pei Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dongguo Li
- School of Biomedical Engineering, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing, China
- *Correspondence: Dongguo Li,
| |
Collapse
|
5
|
Peters NA, Constantinides A, Ubink I, van Kuik J, Bloemendal HJ, van Dodewaard JM, Brink MA, Schwartz TP, Lolkema MP, Lacle MM, Moons LM, Geesing J, van Grevenstein WM, Roodhart JML, Koopman M, Elias SG, Borel Rinkes IH, Kranenburg O. Consensus molecular subtype 4 (CMS4)-targeted therapy in primary colon cancer: A proof-of-concept study. Front Oncol 2022; 12:969855. [PMID: 36147916 PMCID: PMC9486194 DOI: 10.3389/fonc.2022.969855] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundMesenchymal Consensus Molecular Subtype 4 (CMS4) colon cancer is associated with poor prognosis and therapy resistance. In this proof-of-concept study, we assessed whether a rationally chosen drug could mitigate the distinguishing molecular features of primary CMS4 colon cancer.MethodsIn the ImPACCT trial, informed consent was obtained for molecular subtyping at initial diagnosis of colon cancer using a validated RT-qPCR CMS4-test on three biopsies per tumor (Phase-1, n=69 patients), and for neoadjuvant CMS4-targeting therapy with imatinib (Phase-2, n=5). Pre- and post-treatment tumor biopsies were analyzed by RNA-sequencing and immunohistochemistry. Imatinib-induced gene expression changes were associated with molecular subtypes and survival in an independent cohort of 3232 primary colon cancer.ResultsThe CMS4-test classified 52/172 biopsies as CMS4 (30%). Five patients consented to imatinib treatment prior to surgery, yielding 15 pre- and 15 post-treatment samples for molecular analysis. Imatinib treatment caused significant suppression of mesenchymal genes and upregulation of genes encoding epithelial junctions. The gene expression changes induced by imatinib were associated with improved survival and a shift from CMS4 to CMS2.ConclusionImatinib may have value as a CMS-switching drug in primary colon cancer and induces a gene expression program that is associated with improved survival.
Collapse
Affiliation(s)
- Niek A. Peters
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Alexander Constantinides
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Inge Ubink
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Joyce van Kuik
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Haiko J. Bloemendal
- Department of Internal Medicine, Meander Medical Center, Amersfoort, Netherlands
- Department of Internal Medicine/Oncology, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | | | - Menno A. Brink
- Department of Gastroenterology, Meander Medical Center, Amersfoort, Netherlands
| | - Thijs P. Schwartz
- Department of Gastroenterology, Meander Medical Center, Amersfoort, Netherlands
| | | | - Miangela M. Lacle
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Leon M. Moons
- Department of Gastroenterology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Joost Geesing
- Department of Gastroenterology, Diakonessenhuis, Utrecht, Netherlands
| | - Wilhelmina M.U. van Grevenstein
- Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jeanine M. L. Roodhart
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Sjoerd G. Elias
- Julius Centre for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Inne H.M. Borel Rinkes
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- *Correspondence: Inne H.M. Borel Rinkes, ; Onno Kranenburg,
| | - Onno Kranenburg
- Lab Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- *Correspondence: Inne H.M. Borel Rinkes, ; Onno Kranenburg,
| |
Collapse
|
6
|
Ganguli A, Mostafa A, Saavedra C, Kim Y, Le P, Faramarzi V, Feathers RW, Berger J, Ramos-Cruz KP, Adeniba O, Diaz GJP, Drnevich J, Wright CL, Hernandez AG, Lin W, Smith AM, Kosari F, Vasmatzis G, Anastasiadis PZ, Bashir R. Three-dimensional microscale hanging drop arrays with geometric control for drug screening and live tissue imaging. SCIENCE ADVANCES 2021; 7:7/17/eabc1323. [PMID: 33893093 PMCID: PMC8064630 DOI: 10.1126/sciadv.abc1323] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 03/05/2021] [Indexed: 05/09/2023]
Abstract
Existing three-dimensional (3D) culture techniques are limited by trade-offs between throughput, capacity for high-resolution imaging in living state, and geometric control. Here, we introduce a modular microscale hanging drop culture where simple design elements allow high replicates for drug screening, direct on-chip real-time or high-resolution confocal microscopy, and geometric control in 3D. Thousands of spheroids can be formed on our microchip in a single step and without any selective pressure from specific matrices. Microchip cultures from human LN229 glioblastoma and patient-derived mouse xenograft cells retained genomic alterations of originating tumors based on mate pair sequencing. We measured response to drugs over time with real-time microscopy on-chip. Last, by engineering droplets to form predetermined geometric shapes, we were able to manipulate the geometry of cultured cell masses. These outcomes can enable broad applications in advancing personalized medicine for cancer and drug discovery, tissue engineering, and stem cell research.
Collapse
Affiliation(s)
- A Ganguli
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - A Mostafa
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - C Saavedra
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Y Kim
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - P Le
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - V Faramarzi
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - R W Feathers
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - J Berger
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - K P Ramos-Cruz
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - O Adeniba
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - G J Pagan Diaz
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - J Drnevich
- High-Performance Biological Computing, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - C L Wright
- DNA Services Lab, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - A G Hernandez
- DNA Services Lab, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - W Lin
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - A M Smith
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, Urbana, IL 61820, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - F Kosari
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - G Vasmatzis
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - P Z Anastasiadis
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA.
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - R Bashir
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, Urbana, IL 61820, USA
| |
Collapse
|
7
|
Combined crizotinib and endocrine drugs inhibit proliferation, migration, and colony formation of breast cancer cells via downregulation of MET and estrogen receptor. Med Oncol 2021; 38:8. [DOI: 10.1007/s12032-021-01458-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/01/2021] [Indexed: 02/07/2023]
|
8
|
Huelse J, Fridlyand D, Earp S, DeRyckere D, Graham DK. MERTK in cancer therapy: Targeting the receptor tyrosine kinase in tumor cells and the immune system. Pharmacol Ther 2020; 213:107577. [PMID: 32417270 PMCID: PMC9847360 DOI: 10.1016/j.pharmthera.2020.107577] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The receptor tyrosine kinase MERTK is aberrantly expressed in numerous human malignancies, and is a novel target in cancer therapeutics. Physiologic roles of MERTK include regulation of tissue homeostasis and repair, innate immune control, and platelet aggregation. However, aberrant expression in a wide range of liquid and solid malignancies promotes neoplasia via growth factor independence, cell cycle progression, proliferation and tumor growth, resistance to apoptosis, and promotion of tumor metastases. Additionally, MERTK signaling contributes to an immunosuppressive tumor microenvironment via induction of an anti-inflammatory cytokine profile and regulation of the PD-1 axis, as well as regulation of macrophage, myeloid-derived suppressor cell, natural killer cell and T cell functions. Various MERTK-directed therapies are in preclinical development, and clinical trials are underway. In this review we discuss MERTK inhibition as an emerging strategy for cancer therapy, focusing on MERTK expression and function in neoplasia and its role in mediating resistance to cytotoxic and targeted therapies as well as in suppressing anti-tumor immunity. Additionally, we review preclinical and clinical pharmacological strategies to target MERTK.
Collapse
Affiliation(s)
- Justus Huelse
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Diana Fridlyand
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Shelton Earp
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Douglas K. Graham
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Georgia
| |
Collapse
|
9
|
Huelse JM, Fridlyand DM, Earp S, DeRyckere D, Graham DK. MERTK in cancer therapy: Targeting the receptor tyrosine kinase in tumor cells and the immune system. Pharmacol Ther 2020. [PMID: 32417270 DOI: 10.1016/j.pharmthera.2020.107577107577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The receptor tyrosine kinase MERTK is aberrantly expressed in numerous human malignancies, and is a novel target in cancer therapeutics. Physiologic roles of MERTK include regulation of tissue homeostasis and repair, innate immune control, and platelet aggregation. However, aberrant expression in a wide range of liquid and solid malignancies promotes neoplasia via growth factor independence, cell cycle progression, proliferation and tumor growth, resistance to apoptosis, and promotion of tumor metastases. Additionally, MERTK signaling contributes to an immunosuppressive tumor microenvironment via induction of an anti-inflammatory cytokine profile and regulation of the PD-1 axis, as well as regulation of macrophage, myeloid-derived suppressor cell, natural killer cell and T cell functions. Various MERTK-directed therapies are in preclinical development, and clinical trials are underway. In this review we discuss MERTK inhibition as an emerging strategy for cancer therapy, focusing on MERTK expression and function in neoplasia and its role in mediating resistance to cytotoxic and targeted therapies as well as in suppressing anti-tumor immunity. Additionally, we review preclinical and clinical pharmacological strategies to target MERTK.
Collapse
Affiliation(s)
- Justus M Huelse
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Diana M Fridlyand
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Shelton Earp
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, USA.
| |
Collapse
|
10
|
A cancer drug atlas enables synergistic targeting of independent drug vulnerabilities. Nat Commun 2020; 11:2935. [PMID: 32523045 PMCID: PMC7287046 DOI: 10.1038/s41467-020-16735-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
Personalized cancer treatments using combinations of drugs with a synergistic effect is attractive but proves to be highly challenging. Here we present an approach to uncover the efficacy of drug combinations based on the analysis of mono-drug effects. For this we used dose-response data from pharmacogenomic encyclopedias and represent these as a drug atlas. The drug atlas represents the relations between drug effects and allows to identify independent processes for which the tumor might be particularly vulnerable when attacked by two drugs. Our approach enables the prediction of combination-therapy which can be linked to tumor-driving mutations. By using this strategy, we can uncover potential effective drug combinations on a pan-cancer scale. Predicted synergies are provided and have been validated in glioblastoma, breast cancer, melanoma and leukemia mouse-models, resulting in therapeutic synergy in 75% of the tested models. This indicates that we can accurately predict effective drug combinations with translational value.
Collapse
|
11
|
Cruickshanks N, Zhang Y, Hine S, Gibert M, Yuan F, Oxford M, Grello C, Pahuski M, Dube C, Guessous F, Wang B, Deveau C, Saoud K, Gallagher I, Wulfkuhle J, Schiff D, Phan S, Petricoin E, Abounader R. Discovery and Therapeutic Exploitation of Mechanisms of Resistance to MET Inhibitors in Glioblastoma. Clin Cancer Res 2018; 25:663-673. [PMID: 30201763 DOI: 10.1158/1078-0432.ccr-18-0926] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/13/2018] [Accepted: 09/05/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE Glioblastoma (GBM) is the most common and most lethal primary malignant brain tumor. The receptor tyrosine kinase MET is frequently upregulated or overactivated in GBM. Although clinically applicable MET inhibitors have been developed, resistance to single modality anti-MET drugs frequently occurs, rendering these agents ineffective. We aimed to determine the mechanisms of MET inhibitor resistance in GBM and use the acquired information to develop novel therapeutic approaches to overcome resistance.Experimental Design: We investigated two clinically applicable MET inhibitors: crizotinib, an ATP-competitive small molecule inhibitor of MET, and onartuzumab, a monovalent monoclonal antibody that binds to the extracellular domain of the MET receptor. We developed new MET inhibitor-resistant cells lines and animal models and used reverse phase protein arrays (RPPA) and functional assays to uncover the compensatory pathways in MET inhibitor-resistant GBM. RESULTS We identified critical proteins that were altered in MET inhibitor-resistant GBM including mTOR, FGFR1, EGFR, STAT3, and COX-2. Simultaneous inhibition of MET and one of these upregulated proteins led to increased cell death and inhibition of cell proliferation in resistant cells compared with either agent alone. In addition, in vivo treatment of mice bearing MET-resistant orthotopic xenografts with COX-2 or FGFR pharmacological inhibitors in combination with MET inhibitor restored sensitivity to MET inhibition and significantly inhibited tumor growth. CONCLUSIONS These data uncover the molecular basis of adaptive resistance to MET inhibitors and identify new FDA-approved multidrug therapeutic combinations that can overcome resistance.
Collapse
Affiliation(s)
- Nichola Cruickshanks
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Ying Zhang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Sarah Hine
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Myron Gibert
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Fang Yuan
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Madison Oxford
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Cassandra Grello
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Mary Pahuski
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Collin Dube
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Fadila Guessous
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia.,University Mohammed 6 for Health Sciences, Casablanca, Morocco
| | - Baomin Wang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Ciana Deveau
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Karim Saoud
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Isela Gallagher
- George Mason University Center for Applied Proteomics and Molecular Medicine, Manassas, Virginia
| | - Julia Wulfkuhle
- George Mason University Center for Applied Proteomics and Molecular Medicine, Manassas, Virginia
| | - David Schiff
- Department of Neurology, University of Virginia, Charlottesville, Virginia
| | - See Phan
- Genentech Inc. South San Francisco, California
| | - Emanuel Petricoin
- George Mason University Center for Applied Proteomics and Molecular Medicine, Manassas, Virginia
| | - Roger Abounader
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia. .,Department of Neurology, University of Virginia, Charlottesville, Virginia.,The Cancer Center, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
12
|
Combinatorial Drug Testing in 3D Microtumors Derived from GBM Patient-Derived Xenografts Reveals Cytotoxic Synergy in Pharmacokinomics-informed Pathway Interactions. Sci Rep 2018; 8:8412. [PMID: 29849102 PMCID: PMC5976646 DOI: 10.1038/s41598-018-26840-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/15/2018] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most common form of primary malignant brain cancer in adults, is a devastating disease for which effective treatment has remained elusive for over 75 years. One reason for the minimal progress during this time is the lack of accurate preclinical models to represent the patient's tumor's in vivo environment, causing a disconnect in drug therapy effectiveness between the laboratory and clinic. While patient-derived xenografts (PDX's or xenolines) are excellent human tumor representations, they are not amenable to high throughput testing. Therefore, we developed a miniaturized xenoline system (microtumors) for drug testing. Nineteen GBM xenolines were profiled for global kinase (kinomic) activity revealing actionable kinase targets associated with intracranial tumor growth rate. Kinase inhibitors for these targets (WP1066, selumetinib, crizotinib, and cediranib) were selected for single and combination therapy using a fully human-derived three-dimensional (3D) microtumor model of GBM xenoline cells embedded in HuBiogel for subsequent molecular and phenotype assays. GBM microtumors closely resembled orthotopically-implanted tumors based on immunohistochemical analysis and displayed kinomic and morphological diversity. Drug response testing could be reproducibly performed in a 96-well format identifying several synergistic combinations. Our findings indicate that 3D microtumors can provide a suitable high-throughput model for combination drug testing.
Collapse
|
13
|
Recent and future advances in anticancer drug delivery: an interview with Khaled Greish. Ther Deliv 2018; 9:409-412. [PMID: 29722635 DOI: 10.4155/tde-2018-0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Khaled Greish speaks to Hannah Makin, Commissioning Editor: Khaled Greish is Associate Professor of Molecular Medicine, and head of the Nano-research unit, at Princes Al-Jawhara Center, Arabian Gulf University, Kingdom of Bahrain. His previous appointments included Senior lecturer of Pharmacology at the University of Otago, New Zealand, and Assistant Professor of Pharmaceutical Chemistry at University of Utah (UT, USA). He has published >70 peer reviewed papers, and ten book chapters in the field of targeted anticancer drug delivery. Controlled Release Society (CRS) awarded him the CRS Postdoctoral Achievement Award in 2008 and in 2010; he was elected as member of the CRS College of Fellows. In recognition of his research, University of Otago awarded him "Early Career Awards for Distinction in Research" in 2014. His research focuses on nanomedicine, tumor vascular biology and anticancer drug discovery/development.
Collapse
|
14
|
Kim W, Youn H, Lee S, Kim E, Kim D, Sub Lee J, Lee JM, Youn B. RNF138-mediated ubiquitination of rpS3 is required for resistance of glioblastoma cells to radiation-induced apoptosis. Exp Mol Med 2018; 50:e434. [PMID: 29371697 PMCID: PMC5799804 DOI: 10.1038/emm.2017.247] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/10/2017] [Accepted: 07/23/2017] [Indexed: 02/06/2023] Open
Abstract
An interaction between ribosomal protein S3 (rpS3) and nuclear factor kappa B or macrophage migration inhibitory factor in non-small-cell lung cancer is responsible for radioresistance. However, the role of rpS3 in glioblastoma (GBM) has not been investigated to date. Here we found that in irradiated GBM cells, rpS3 translocated into the nucleus and was subsequently ubiquitinated by ring finger protein 138 (RNF138). Ubiquitin-dependent degradation of rpS3 consequently led to radioresistance in GBM cells. To elucidate the apoptotic role of rpS3, we analyzed the interactome of rpS3 in ΔRNF138 GBM cells. Nuclear rpS3 interacted with DNA damage inducible transcript 3 (DDIT3), leading to DDIT3-induced apoptosis in irradiated ΔRNF138 GBM cells. These results were confirmed using in vivo orthotopic xenograft models and GBM patient tissues. This study aims to clarify the role of RNF138 in GBM cells and demonstrate that rpS3 may be a promising substrate of RNF138 for the induction of GBM radioresistance, indicating RNF138 as a potential target for GBM therapy.
Collapse
Affiliation(s)
- Wanyeon Kim
- Department of Biological Sciences, Pusan National University, Busan, Republic of Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, Republic of Korea
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - EunGi Kim
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - Daehoon Kim
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - Jung Sub Lee
- Department of Orthopaedic Surgery, Medical Research Institute, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Jae-Myung Lee
- Department of Naval Architecture and Ocean Engineering, Pusan National University, Busan, Republic of Korea
| | - BuHyun Youn
- Department of Biological Sciences, Pusan National University, Busan, Republic of Korea
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
15
|
He Y, Zhou L, Gao S, Yin T, Tu Y, Rayford R, Wang X, Hu M. Development and validation of a sensitive LC–MS/MS method for simultaneous determination of eight tyrosine kinase inhibitors and its application in mice pharmacokinetic studies. J Pharm Biomed Anal 2018; 148:65-72. [DOI: 10.1016/j.jpba.2017.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/15/2017] [Accepted: 09/08/2017] [Indexed: 11/26/2022]
|
16
|
Greish K, Jasim A, Parayath N, Abdelghany S, Alkhateeb A, Taurin S, Nehoff H. Micellar formulations of Crizotinib and Dasatinib in the management of glioblastoma multiforme. J Drug Target 2017; 26:692-708. [PMID: 29251531 DOI: 10.1080/1061186x.2017.1419357] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) defies the currently practiced management of radiotherapy, chemotherapy and surgery and hence, it is associated with a high fatality rate with a median survival of 14.6 months. In our previous work investigating different tyrosine kinase inhibitors (TKIs), we established that a combination of Crizotinib and Dasatinib exerted the most potent effect on different GBM cell lines. In this work, to improve targeted therapy at the site of the tumour and avoid systemic toxicity, we exploited the enhanced permeability and retention effect by designing micellar formulations of these two TKIs. Crizotinib and Dasatinib were successfully encapsulated in poly(styrene-co-maleic acid) (SMA) micelles which were then evaluated for their physicochemical characteristics, anti-proliferative effect, mode of cell death, efficacy in spheroid models, effect on cell signalling, antiangiogenic potential and in vivo anticancer activity. Our results showed that this combination had induced a potent anti-proliferative effect in four GBM cell lines grown as a monolayer and as a spheroid. The combination was also efficacious in in vitro models of angiogenesis and vascular mimicry. In vivo data showed the enhanced activity of the micellar TKIs compared to free drugs. In conclusion, we proved that micellar formulations of Crizotinib and Dasatinib carry promising in vitro and in vivo efficacy that warrant further investigation.
Collapse
Affiliation(s)
- Khaled Greish
- a College of Medicine and Medical Sciences, Department of Molecular Medicine, and Nanomedicine Unit , Princess Al-Jawhara Center for Molecular Medicine, Arabian Gulf University , Manama , Kingdom of Bahrain
| | - Anfal Jasim
- a College of Medicine and Medical Sciences, Department of Molecular Medicine, and Nanomedicine Unit , Princess Al-Jawhara Center for Molecular Medicine, Arabian Gulf University , Manama , Kingdom of Bahrain
| | - Neha Parayath
- b Department of Pharmaceutical Sciences , Northeastern University , Boston , MA , USA
| | - Sara Abdelghany
- a College of Medicine and Medical Sciences, Department of Molecular Medicine, and Nanomedicine Unit , Princess Al-Jawhara Center for Molecular Medicine, Arabian Gulf University , Manama , Kingdom of Bahrain
| | - Ali Alkhateeb
- a College of Medicine and Medical Sciences, Department of Molecular Medicine, and Nanomedicine Unit , Princess Al-Jawhara Center for Molecular Medicine, Arabian Gulf University , Manama , Kingdom of Bahrain
| | - Sebastien Taurin
- c Department of Obstetrics and Gynecology , University of Utah , Salt Lake City , UT , USA
| | - Hayley Nehoff
- d Department of Pharmacology and Toxicology , University of Otago , Dunedin , New Zealand
| |
Collapse
|
17
|
Phase I study of the combination of crizotinib (as a MET inhibitor) and dasatinib (as a c-SRC inhibitor) in patients with advanced cancer. Invest New Drugs 2017; 36:416-423. [PMID: 29047029 DOI: 10.1007/s10637-017-0513-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/20/2017] [Indexed: 01/19/2023]
Abstract
Background Both MET and c-SRC are important mediators of cancer progression and there is cross talk between the two molecules. Preclinical studies have demonstrated combination of MET and c-SRC inhibitors is effective in multiple cancer types. Methods We analyzed the safety and efficacy of administering a c-SRC inhibitor (dasatinib) in combination with a MET inhibitor (crizotinib) in a two-arm concurrent phase I study. Arm A consisted of crizotinib fixed at 250 mg twice per day with escalation of dasatinib. Arm B consisted of dasatinib fixed at 140 mg daily with escalation of crizotinib. Endpoints included dose-limiting toxicities (DLTs), recommended phase II dose (RP2D), and response (RECIST 1.1). Results We enrolled 61 patients (arm A: 31, arm B: 30). The most common cancers were sarcoma (21%) and prostate cancer (16%). In Arm A, at dose level 2 (DL2), 40% (2/5) experienced DLTs. In the expanded DL1, 21% (4/19) experienced DLTs (all grade 3). In Arm B, at DL2, 50% (2/4) experienced DLTs. In the expanded DL1, 22% (4/18) experienced DLTs (all grade 3). RP2D was determined to be arm A, DL1 (250 mg crizotinib orally twice per day plus 50 mg dasatinib orally daily). Partial response (N = 1) and stable disease for ≥6 months (N = 3) were seen. Conclusions The combination of crizotinib and dasatinib is safe to administer but tolerability is limited given the high rate of adverse events. Responses and durable stable disease were limited. Further precision therapy approach using this specific combination may be difficult given the toxicity.
Collapse
|
18
|
Role and Therapeutic Targeting of the HGF/MET Pathway in Glioblastoma. Cancers (Basel) 2017; 9:cancers9070087. [PMID: 28696366 PMCID: PMC5532623 DOI: 10.3390/cancers9070087] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 06/29/2017] [Accepted: 07/06/2017] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GBM) is a lethal brain tumor with dismal prognosis. Current therapeutic options, consisting of surgery, chemotherapy and radiation, have only served to marginally increase patient survival. Receptor tyrosine kinases (RTKs) are dysregulated in approximately 90% of GBM; attributed to this, research has focused on inhibiting RTKs as a novel and effective therapy for GBM. Overexpression of RTK mesenchymal epithelial transition (MET), and its ligand, hepatocyte growth factor (HGF), in GBM highlights a promising new therapeutic target. This review will discuss the role of MET in cell cycle regulation, cell proliferation, evasion of apoptosis, cell migration and invasion, angiogenesis and therapeutic resistance in GBM. It will also discuss the modes of deregulation of HGF/MET and their regulation by microRNAs. As the HGF/MET pathway is a vital regulator of multiple pro-survival pathways, efforts and strategies for its exploitation for GBM therapy are also described.
Collapse
|
19
|
Huang XX, Xie FF, Hou LJ, Chen XX, Ou RY, Yu JT, Qiu JG, Zhang WJ, Jiang QW, Yang Y, Zheng DW, Chen Y, Huang JR, Wang K, Wei MN, Li WF, Shi Z, Yan XJ. Crizotinib synergizes with cisplatin in preclinical models of ovarian cancer. Am J Transl Res 2017; 9:1667-1679. [PMID: 28469773 PMCID: PMC5411916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/04/2017] [Indexed: 06/07/2023]
Abstract
Crizotinib, a small molecule inhibitor of anaplastic lymphoma kinase (ALK), c-ros oncogene 1 (ROS1) and c-MET (also called MET or hepatocyte growth factor receptor), has been approved by the Food and Drug Administration for the treatment of patients with advanced non-small cell lung cancer whose tumors have rearrangements in the ALK or ROS1 gene. However, the anticancer effect of crizotinib on ovarian cancer is still unclear. In this study, our data show that crizotinib can actively induce cell growth inhibition, cell cycle arrest at G2/M phase and apoptosis with the decreasing phosphorylation of the downstream signaling effectors AKT and ERK in human ovarian cancer cells. Crizotinib also increases the intracellular reactive oxidative species (ROS) levels, and pretreating with ROS scavenger N-acety-L-cysteine partially reverses crizotinib-induced apoptosis. Moreover, crizotinib can synergistically inhibit ovarian cancer cells growth in vitro and in vivo when combines with cisplatin. Altogether, crizotinib potently potentiates the activity of cisplatin in ovarian cancer, suggesting the synergistic effect of crizotinib and cisplatin may be valuable for ovarian cancer patients' treatment.
Collapse
Affiliation(s)
- Xiao-Xiu Huang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Feng-Feng Xie
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Li-Jiao Hou
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Xiu-Xiu Chen
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Rong-Ying Ou
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Jiang-Tao Yu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Jian-Ge Qiu
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Wen-Ji Zhang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Qi-Wei Jiang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Yang Yang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Di-Wei Zheng
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Yao Chen
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Jia-Rong Huang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Kun Wang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Meng-Ning Wei
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Wen-Feng Li
- Department of Chemoradiotherapy, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Zhi Shi
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan UniversityGuangzhou 510632, Guangdong, China
| | - Xiao-Jian Yan
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| |
Collapse
|
20
|
Zhang Z, Song X, Tian H, Miao Y, Feng X, Li Y, Wang H. MicroRNA-137 inhibits growth of glioblastoma through EGFR suppression. Am J Transl Res 2017; 9:1492-1499. [PMID: 28386374 PMCID: PMC5376039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 02/08/2017] [Indexed: 06/07/2023]
Abstract
Aberrant expression of certain microRNAs (miRNAs) has been shown to contribute to the development of Glioblastoma multiforme (GBM). However, the involvement of miR-137 in the carcinogenesis of GBM has not been reported. Here, we showed that miR-137 levels in GBM tissues were significantly lower than the paired normal brain tissue in patients' specimens. Moreover, low miR-137 levels in GBM tissue were associated with poor prognosis. In vitro, overexpression of miR-137 decreased GBM cell growth and increased cell apoptosis, while depletion of miR-137 enhanced cell growth and decreased cell apoptosis. Combined bioinformatics analysis and dual luciferase reporter assay showed that miR-137 may target the 3'-UTR of the epidermal growth factor receptor (EGFR) to reduce its protein translation, resulting in suppression of EGFR signaling in GBM cells. Together, our data suggest that reduction in miR-137 levels in GBM tissues may increase cell growth and decrease cell apoptosis, possibly through suppression of EGFR.
Collapse
Affiliation(s)
- Zhenxing Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou 121001, China
| | - Xiaofeng Song
- Department of Histology and Embryology, Jinzhou Medical UniversityJinzhou 121001, China
| | - He Tian
- Department of Histology and Embryology, Jinzhou Medical UniversityJinzhou 121001, China
| | - Ye Miao
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou 121001, China
| | - Xu Feng
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou 121001, China
| | - Yang Li
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou 121001, China
| | - Honglei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou 121001, China
| |
Collapse
|
21
|
Haddad J, Slika S, Mahfouz R. Epidermal growth factor receptor (EGFR) in the era of Precision Medicine: The tale of a perfect example of targeted therapy. A review. Meta Gene 2017. [DOI: 10.1016/j.mgene.2016.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
22
|
Abstract
INTRODUCTION Despite substantial improvements in standards of care, the most common aggressive pediatric and adult high-grade gliomas (HGG) carry uniformly fatal diagnoses due to unique treatment limitations, high recurrence rates and the absence of effective treatments following recurrence. Recent advancements in our understanding of the pathophysiology, genetics and epigenetics as well as mechanisms of immune surveillance during gliomagenesis have created new knowledge to design more effective and target-directed therapies to improve patient outcomes. AREAS COVERED In this review, the authors discuss the critical genetic, epigenetic and immunologic aberrations found in gliomas that appear rational and promising for therapeutic developments in the presence and future. The current state of the latest therapeutic developments including tumor-specific targeted drug therapies, metabolic targeting, epigenetic modulation and immunotherapy are summarized and suggestions for future directions are offered. Furthermore, they highlight contemporary issues related to the clinical development, such as challenges in clinical trials and toxicities. EXPERT OPINION The commitment to understanding the process of gliomagenesis has created a catalogue of aberrations that depict multiple mechanisms underlying this disease, many of which are suitable to therapeutic inhibition and are currently tested in clinical trials. Thus, future treatment endeavors will employ multiple treatment modalities that target disparate tumor characteristics personalized to the patient's individual tumor.
Collapse
Affiliation(s)
- Verena Staedtke
- a Department of Neurology , Johns Hopkins Medical Institutions , Baltimore , MD , USA
| | - Ren-Yuan Bai
- b Department of Neurosurgery , Johns Hopkins Medical Institutions , Baltimore , MD , USA
| | - John Laterra
- a Department of Neurology , Johns Hopkins Medical Institutions , Baltimore , MD , USA.,c Department of Oncology , Johns Hopkins Medical Institutions , Baltimore , MD , USA.,d Department of Neuroscience , Johns Hopkins Medical Institutions , Baltimore , MD , USA
| |
Collapse
|