1
|
Saeedi BJ, Carr HE, Higgins PDR, Steiner CA. AXL: A novel therapeutic target in IBD. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:141-157. [PMID: 39521598 DOI: 10.1016/bs.apha.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Inflammatory bowel diseases (IBD) and their sequela (colitis-associate carcinoma and fibrostenotic complications) remain a significant clinical challenge and novel therapeutic targets are desperately needed. AXL, a receptor tyrosine kinase, has been implicated in myriad cellular functions central to the pathogenesis of IBD. These include facilitating epithelial-to-mesenchymal transition, dampening of Toll-like receptor and natural killer cell mediated immune responses, driving proliferation, and propagating fibrogenic signaling. The vast majority of preclinical research on AXL has focused on its role in cancer. As such, pharmacologic AXL inhibitors are currently in clinical trials, but the indications remain limited to malignancy. In this chapter, we summarize the current preclinical data of AXL in IBD, colitis associated carcinoma, and fibrostenotic disease, and highlight its potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Bejan J Saeedi
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Mucosal Inflammation Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States.
| | - Hannah E Carr
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Peter D R Higgins
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI, United States
| | - Calen A Steiner
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Mucosal Inflammation Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
2
|
Behrmann CA, Ennis KN, Sarma P, Wetzel C, Clark NA, Von Handorf KM, Vallabhapurapu S, Andreani C, Reigle J, Scaglioni PP, Meller J, Czyzyk-Krzeska MF, Kendler A, Qi X, Sarkaria JN, Medvedovic M, Sengupta S, Dasgupta B, Plas DR. Coordinated Targeting of S6K1/2 and AXL Disrupts Pyrimidine Biosynthesis in PTEN-Deficient Glioblastoma. CANCER RESEARCH COMMUNICATIONS 2024; 4:2215-2227. [PMID: 39087397 PMCID: PMC11342319 DOI: 10.1158/2767-9764.crc-23-0631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/20/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Intrinsic resistance to targeted therapeutics in PTEN-deficient glioblastoma (GBM) is mediated by redundant signaling networks that sustain critical metabolic functions. Here, we demonstrate that coordinated inhibition of the ribosomal protein S6 kinase 1 (S6K1) and the receptor tyrosine kinase AXL using LY-2584702 and BMS-777607 can overcome network redundancy to reduce GBM tumor growth. This combination of S6K1 and AXL inhibition suppressed glucose flux to pyrimidine biosynthesis. Genetic inactivation studies to map the signaling network indicated that both S6K1 and S6K2 transmit growth signals in PTEN-deficient GBM. Kinome-wide ATP binding analysis in inhibitor-treated cells revealed that LY-2584702 directly inhibited S6K1, and substrate phosphorylation studies showed that BMS-777607 inactivation of upstream AXL collaborated to reduce S6K2-mediated signal transduction. Thus, combination targeting of S6K1 and AXL provides a kinase-directed therapeutic approach that circumvents signal transduction redundancy to interrupt metabolic function and reduce growth of PTEN-deficient GBM. SIGNIFICANCE Therapy for glioblastoma would be advanced by incorporating molecularly targeted kinase-directed agents, similar to standard of care strategies in other tumor types. Here, we identify a kinase targeting approach to inhibit the metabolism and growth of glioblastoma.
Collapse
Affiliation(s)
- Catherine A. Behrmann
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Kelli N. Ennis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Pranjal Sarma
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Collin Wetzel
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Nicholas A. Clark
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Kate M. Von Handorf
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Subrahmanya Vallabhapurapu
- Division of Hematology-Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
- UC Brain Tumor Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Cristina Andreani
- Division of Hematology-Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - James Reigle
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Pier Paolo Scaglioni
- Division of Hematology-Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Jarek Meller
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Maria F. Czyzyk-Krzeska
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
- Department of Veterans Affairs, Cincinnati Veteran Affairs Medical Center, Cincinnati, Ohio.
- Department of Pharmacology and Systems Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Ady Kendler
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Xiaoyang Qi
- Division of Hematology-Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
- UC Brain Tumor Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Mario Medvedovic
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - Soma Sengupta
- UC Brain Tumor Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.
- Departments of Neurology and Neurosurgery, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.
| | - Biplab Dasgupta
- UC Brain Tumor Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio.
| | - David R. Plas
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
- UC Brain Tumor Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
3
|
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
|
4
|
Li F, Xu L, Li C, Hu F, Su Y. Immunological role of Gas6/TAM signaling in hemostasis and thrombosis. Thromb Res 2024; 238:161-171. [PMID: 38723521 DOI: 10.1016/j.thromres.2024.05.002] [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: 03/13/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/21/2024]
Abstract
The immune system is an emerging regulator of hemostasis and thrombosis. The concept of immunothrombosis redefines the relationship between coagulation and immunomodulation, and the Gas6/Tyro3-Axl-MerTK (TAM) signaling pathway builds the bridge across them. During coagulation, Gas6/TAM signaling pathway not only activates platelets, but also promotes thrombosis through endothelial cells and vascular smooth muscle cells involved in inflammatory responses. Thrombosis appears to be a common result of a Gas6/TAM signaling pathway-mediated immune dysregulation. TAM TK and its ligands have been found to be involved in coagulation through the PI3K/AKT or JAK/STAT pathway in various systemic diseases, providing new perspectives in the understanding of immunothrombosis. Gas6/TAM signaling pathway serves as a breakthrough target for novel therapeutic strategies to improve disease management. Many preclinical and clinical studies of TAM receptor inhibitors are in process, confirming the pivotal role of Gas6/TAM signaling pathway in immunothrombosis. Therapeutics targeting the TAM receptor show potential both in anticoagulation management and immunotherapy. Here, we review the immunological functions of the Gas6/TAM signaling pathway in coagulation and its multiple mechanisms in diseases identified to date, and discuss the new clinical strategies that may generated by these roles.
Collapse
Affiliation(s)
- Fanshu Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Liling Xu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| | - Chun Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Yin Su
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China; Peking University People's Hospital, Qingdao, China
| |
Collapse
|
5
|
Fang F, Dai Y, Wang H, Ji Y, Liang X, Peng X, Li J, Zhao Y, Li C, Wang D, Li Y, Zhang D, Zhang D, Geng M, Liu H, Ai J, Zhou Y. Structure-based drug discovery of novel fused-pyrazolone carboxamide derivatives as potent and selective AXL inhibitors. Acta Pharm Sin B 2023; 13:4918-4933. [PMID: 38045061 PMCID: PMC10692477 DOI: 10.1016/j.apsb.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/02/2023] [Accepted: 09/25/2023] [Indexed: 12/05/2023] Open
Abstract
As a novel and promising antitumor target, AXL plays an important role in tumor growth, metastasis, immunosuppression and drug resistance of various malignancies, which has attracted extensive research interest in recent years. In this study, by employing the structure-based drug design and bioisosterism strategies, we designed and synthesized in total 54 novel AXL inhibitors featuring a fused-pyrazolone carboxamide scaffold, of which up to 20 compounds exhibited excellent AXL kinase and BaF3/TEL-AXL cell viability inhibitions. Notably, compound 59 showed a desirable AXL kinase inhibitory activity (IC50: 3.5 nmol/L) as well as good kinase selectivity, and it effectively blocked the cellular AXL signaling. In turn, compound 59 could potently inhibit BaF3/TEL-AXL cell viability (IC50: 1.5 nmol/L) and significantly suppress GAS6/AXL-mediated cancer cell invasion, migration and wound healing at the nanomolar level. More importantly, compound 59 oral administration showed good pharmacokinetic profile and in vivo antitumor efficiency, in which we observed significant AXL phosphorylation suppression, and its antitumor efficacy at 20 mg/kg (qd) was comparable to that of BGB324 at 50 mg/kg (bid), the most advanced AXL inhibitor. Taken together, this work provided a valuable lead compound as a potential AXL inhibitor for the further antitumor drug development.
Collapse
Affiliation(s)
| | - Yang Dai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hao Wang
- Drug Discovery & Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yinchun Ji
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xuewu Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xia Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jiyuan Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yangrong Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chunpu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Danyi Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yazhou Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Dong Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Dan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Meiyu Geng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Jing Ai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Yu Zhou
- Drug Discovery & Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| |
Collapse
|
6
|
Vo TTT, Tran Q, Hong Y, Lee H, Cho H, Kim M, Park S, Kim C, Bayarmunkh C, Boldbaatar D, Kwon SH, Park J, Kim SH, Park J. AXL is required for hypoxia-mediated hypoxia-inducible factor-1 alpha function in glioblastoma. Toxicol Res 2023; 39:669-679. [PMID: 37779588 PMCID: PMC10541364 DOI: 10.1007/s43188-023-00195-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/05/2023] [Accepted: 05/24/2023] [Indexed: 10/03/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive type of central nervous system tumor. Molecular targeting may be important when developing efficient GBM treatment strategies. Sequencing of GBMs revealed that the receptor tyrosine kinase (RTK)/RAS/phosphatidylinositol-3-kinase pathway was altered in 88% of samples. Interestingly, AXL, a member of RTK, was proposed as a promising target in glioma therapy. However, the molecular mechanism of AXL modulation of GBM genesis and proliferation is still unclear. In this study, we investigated the expression and localization of hypoxia-inducible factor-1 alpha (HIF-1α) by AXL in GBM. Both AXL mRNA and protein are overexpressed in GBM. Short-interfering RNA knockdown of AXL in U251-MG cells reduced viability and migration. However, serum withdrawal reduced AXL expression, abolishing the effect on viability. AXL is also involved in hypoxia regulation. In hypoxic conditions, the reduction of AXL decreased the level and nuclear localization of HIF-1α. The co-expression of HIF-1α and AXL was found in human GBM samples but not normal tissue. This finding suggests a mechanism for GBM proliferation and indicates that targeting AXL may be a potential GBM therapeutic. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-023-00195-z.
Collapse
Affiliation(s)
- Thuy-Trang T. Vo
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
| | - Quangdon Tran
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
| | - Youngeun Hong
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
| | - Hyunji Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
| | - Hyeonjeong Cho
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
| | - Minhee Kim
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
| | - Sungjin Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
| | - Chaeyeong Kim
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
| | - Choinyam Bayarmunkh
- Department of Graduate Education, Graduate School, Mongolian National University of Medical Sciences, Ulaanbaatar, 14210 Mongolia
- Department of Physiology, Mongolian National University of Medical Sciences, Ulaanbaatar, 14210 Mongolia
| | - Damdindorj Boldbaatar
- Department of Graduate Education, Graduate School, Mongolian National University of Medical Sciences, Ulaanbaatar, 14210 Mongolia
- Department of Physiology, Mongolian National University of Medical Sciences, Ulaanbaatar, 14210 Mongolia
| | - So Hee Kwon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983 Republic of Korea
| | - Jisoo Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Life Science, Hyehwa Liberal Arts College, LINC Plus Project Group, Daejeon University, Daejeon, 34520 Republic of Korea
| | - Seon-Hwan Kim
- Department of Neurosurgery, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
| | - Jongsun Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
| |
Collapse
|
7
|
Wu Y, Walker JR, Westberg M, Ning L, Monje M, Kirkland TA, Lin MZ, Su Y. Kinase-Modulated Bioluminescent Indicators Enable Noninvasive Imaging of Drug Activity in the Brain. ACS CENTRAL SCIENCE 2023; 9:719-732. [PMID: 37122464 PMCID: PMC10141594 DOI: 10.1021/acscentsci.3c00074] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Indexed: 05/03/2023]
Abstract
Aberrant kinase activity contributes to the pathogenesis of brain cancers, neurodegeneration, and neuropsychiatric diseases, but identifying kinase inhibitors that function in the brain is challenging. Drug levels in blood do not predict efficacy in the brain because the blood-brain barrier prevents entry of most compounds. Rather, assessing kinase inhibition in the brain requires tissue dissection and biochemical analysis, a time-consuming and resource-intensive process. Here, we report kinase-modulated bioluminescent indicators (KiMBIs) for noninvasive longitudinal imaging of drug activity in the brain based on a recently optimized luciferase-luciferin system. We develop an ERK KiMBI to report inhibitors of the Ras-Raf-MEK-ERK pathway, for which no bioluminescent indicators previously existed. ERK KiMBI discriminates between brain-penetrant and nonpenetrant MEK inhibitors, reveals blood-tumor barrier leakiness in xenograft models, and reports MEK inhibitor pharmacodynamics in native brain tissues and intracranial xenografts. Finally, we use ERK KiMBI to screen ERK inhibitors for brain efficacy, identifying temuterkib as a promising brain-active ERK inhibitor, a result not predicted from chemical characteristics alone. Thus, KiMBIs enable the rapid identification and pharmacodynamic characterization of kinase inhibitors suitable for treating brain diseases.
Collapse
Affiliation(s)
- Yan Wu
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
| | - Joel R. Walker
- Promega
Biosciences LLC, San Luis Obispo, California 93401, United States
| | - Michael Westberg
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
- Department
of Chemistry, Aarhus University, Aarhus 8000, Denmark
| | - Lin Ning
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
| | - Michelle Monje
- Department
of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, United States
- Howard Hughes
Medical Institute, Stanford University, Stanford, California 94305, United States
| | - Thomas A. Kirkland
- Promega
Biosciences LLC, San Luis Obispo, California 93401, United States
| | - Michael Z. Lin
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
- Department
of Pediatrics, Stanford University, Stanford, California 94305, United States
- Department
of Chemical and Systems Biology, Stanford
University, Stanford, California 94305, United States
| | - Yichi Su
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
8
|
Two-Front War on Cancer-Targeting TAM Receptors in Solid Tumour Therapy. Cancers (Basel) 2022; 14:cancers14102488. [PMID: 35626092 PMCID: PMC9140196 DOI: 10.3390/cancers14102488] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary In recent years, many studies have shown the importance of TAM kinases in both normal and neoplastic cells. In this review, we present and discuss the role of the TAM family (AXL, MERTK, TYRO3) of receptor tyrosine kinases (RTKs) as a dual target in cancer, due to their intrinsic roles in tumour cell survival, migration, chemoresistance, and their immunosuppressive roles in the tumour microenvironment. This review presents the potential of TAMs as emerging therapeutic targets in cancer treatment, focusing on the distinct structures of TAM receptor tyrosine kinases. We analyse and compare different strategies of TAM inhibition, for a full perspective of current and future battlefields in the war with cancer. Abstract Receptor tyrosine kinases (RTKs) are transmembrane receptors that bind growth factors and cytokines and contain a regulated kinase activity within their cytoplasmic domain. RTKs play an important role in signal transduction in both normal and malignant cells, and their encoding genes belong to the most frequently affected genes in cancer cells. The TAM family proteins (TYRO3, AXL, and MERTK) are involved in diverse biological processes: immune regulation, clearance of apoptotic cells, platelet aggregation, cell proliferation, survival, and migration. Recent studies show that TAMs share overlapping functions in tumorigenesis and suppression of antitumour immunity. MERTK and AXL operate in innate immune cells to suppress inflammatory responses and promote an immunosuppressive tumour microenvironment, while AXL expression correlates with epithelial-to-mesenchymal transition, metastasis, and motility in tumours. Therefore, TAM RTKs represent a dual target in cancer due to their intrinsic roles in tumour cell survival, migration, chemoresistance, and their immunosuppressive roles in the tumour microenvironment (TME). In this review, we discuss the potential of TAMs as emerging therapeutic targets in cancer treatment. We critically assess and compare current approaches to target TAM RTKs in solid tumours and the development of new inhibitors for both extra- and intracellular domains of TAM receptor kinases.
Collapse
|
9
|
Scherschinski L, Prem M, Kremenetskaia I, Tinhofer I, Vajkoczy P, Karbe AG, Onken JS. Regulation of the Receptor Tyrosine Kinase AXL in Response to Therapy and Its Role in Therapy Resistance in Glioblastoma. Int J Mol Sci 2022; 23:ijms23020982. [PMID: 35055167 PMCID: PMC8781963 DOI: 10.3390/ijms23020982] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 01/08/2023] Open
Abstract
The receptor tyrosine kinase AXL (RTK-AXL) is implicated in therapy resistance and tumor progression in glioblastoma multiforme (GBM). Here, we investigated therapy-induced receptor modifications and how endogenous RTK-AXL expression and RTK-AXL inhibition contribute to therapy resistance in GBM. GBM cell lines U118MG and SF126 were exposed to temozolomide (TMZ) and radiation (RTX). Receptor modifications in response to therapy were investigated on protein and mRNA levels. TMZ-resistant and RTK-AXL overexpressing cell lines were exposed to increasing doses of TMZ and RTX, with and without RTK-AXL tyrosine kinase inhibitor (TKI). Colorimetric microtiter (MTT) assay and colony formation assay (CFA) were used to assess cell viability. Results showed that the RTK-AXL shedding product, C-terminal AXL (CT-AXL), rises in response to repeated TMZ doses and under hypoxia, acts as a surrogate marker for radio-resistance. Endogenous RTX-AXL overexpression leads to therapy resistance, whereas combination therapy of TZM and RTX with TKI R428 significantly increases therapeutic effects. This data proves the role of RTK-AXL in acquired and intrinsic therapy resistance. By demonstrating that therapy resistance may be overcome by combining AXL TKI with standard treatments, we have provided a rationale for future study designs investigating AXL TKIs in GBM.
Collapse
Affiliation(s)
- Lea Scherschinski
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Markus Prem
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
- Department of Neurosurgery, Technische Universität Dresden, 01069 Dresden, Germany
| | - Irina Kremenetskaia
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany;
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung–DKTK), Partner Site Berlin, 10115 Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Anna-Gila Karbe
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
| | - Julia Sophie Onken
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.S.); (M.P.); (I.K.); (P.V.); (A.-G.K.)
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung–DKTK), Partner Site Berlin, 10115 Berlin, Germany
- Correspondence: ; Tel.: +49-(0)30-450-660253
| |
Collapse
|
10
|
The Pivotal Immunomodulatory and Anti-Inflammatory Effect of Histone-Lysine N-Methyltransferase in the Glioma Microenvironment: Its Biomarker and Therapy Potentials. Anal Cell Pathol (Amst) 2021; 2021:4907167. [PMID: 34745848 PMCID: PMC8566080 DOI: 10.1155/2021/4907167] [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: 08/28/2021] [Accepted: 10/16/2021] [Indexed: 11/18/2022] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a histone-lysine N-methyltransferase that encrypts a member of the Polycomb group (PcG) family. EZH2 forms a repressive chromatin structure which eventually participates in regulating the development as well as lineage propagation of stem cells and glioma progression. Posttranslational modifications are distinct approaches for the adjusted modification of EZH2 in the development of cancer. The amino acid succession of EZH2 protein makes it appropriate for covalent modifications, like phosphorylation, acetylation, O-GlcNAcylation, methylation, ubiquitination, and sumoylation. The glioma microenvironment is a dynamic component that comprises, besides glioma cells and glioma stem cells, a complex network that comprises diverse cell types like endothelial cells, astrocytes, and microglia as well as stromal components, soluble factors, and the extracellular membrane. EZH2 is well recognized as an essential modulator of cell invasion as well as metastasis in glioma. EZH2 oversecretion was implicated in the malfunction of several fundamental signaling pathways like Wnt/β-catenin signaling, Ras and NF-κB signaling, PI3K/AKT signaling, β-adrenergic receptor signaling, and bone morphogenetic protein as well as NOTCH signaling pathways. EZH2 was more secreted in glioblastoma multiforme than in low-grade gliomas as well as extremely secreted in U251 and U87 human glioma cells. Thus, the blockade of EZH2 expression in glioma could be of therapeutic value for patients with glioma. The suppression of EZH2 gene secretion was capable of reversing temozolomide resistance in patients with glioma. EZH2 is a promising therapeutic as well as prognostic biomarker for the treatment of glioma.
Collapse
|
11
|
Khera L, Lev S. Accelerating AXL targeting for TNBC therapy. Int J Biochem Cell Biol 2021; 139:106057. [PMID: 34403827 DOI: 10.1016/j.biocel.2021.106057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022]
Abstract
The tyrosine kinase receptor AXL of the TAM (TYRO3, AXL and MERTK) family is considered as a promising therapeutic target for different hematological cancers and solid tumors. AXL is involved in multiple pro-tumorigenic processes including cell migration, invasion, epithelial-mesenchymal transition (EMT), and stemness, and recent studies demonstrated its impact on cancer metastasis and drug resistance. Extensive studies on AXL have highlighted its unique characteristics and physiological functions and suggest that targeting of AXL could be beneficial in combination with chemotherapy, radiotherapy, immunotherapy, and targeted therapy. In this mini review, we discuss possible outcomes of AXL targeting either alone or together with other therapeutic agents and emphasize its impact on triple negative breast cancer (TNBC).
Collapse
Affiliation(s)
- Lohit Khera
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
12
|
Therapeutic Targeting of the Gas6/Axl Signaling Pathway in Cancer. Int J Mol Sci 2021; 22:ijms22189953. [PMID: 34576116 PMCID: PMC8469858 DOI: 10.3390/ijms22189953] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 12/14/2022] Open
Abstract
Many signaling pathways are dysregulated in cancer cells and the host tumor microenvironment. Aberrant receptor tyrosine kinase (RTK) pathways promote cancer development, progression, and metastasis. Hence, numerous therapeutic interventions targeting RTKs have been actively pursued. Axl is an RTK that belongs to the Tyro3, Axl, MerTK (TAM) subfamily. Axl binds to a high affinity ligand growth arrest specific 6 (Gas6) that belongs to the vitamin K-dependent family of proteins. The Gas6/Axl signaling pathway has been implicated to promote progression, metastasis, immune evasion, and therapeutic resistance in many cancer types. Therapeutic agents targeting Gas6 and Axl have been developed, and promising results have been observed in both preclinical and clinical settings when such agents are used alone or in combination therapy. This review examines the current state of therapeutics targeting the Gas6/Axl pathway in cancer and discusses Gas6- and Axl-targeting agents that have been evaluated preclinically and clinically.
Collapse
|
13
|
Zdżalik-Bielecka D, Poświata A, Kozik K, Jastrzębski K, Schink KO, Brewińska-Olchowik M, Piwocka K, Stenmark H, Miączyńska M. The GAS6-AXL signaling pathway triggers actin remodeling that drives membrane ruffling, macropinocytosis, and cancer-cell invasion. Proc Natl Acad Sci U S A 2021; 118:e2024596118. [PMID: 34244439 PMCID: PMC8285903 DOI: 10.1073/pnas.2024596118] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AXL, a member of the TAM (TYRO3, AXL, MER) receptor tyrosine kinase family, and its ligand, GAS6, are implicated in oncogenesis and metastasis of many cancer types. However, the exact cellular processes activated by GAS6-AXL remain largely unexplored. Here, we identified an interactome of AXL and revealed its associations with proteins regulating actin dynamics. Consistently, GAS6-mediated AXL activation triggered actin remodeling manifested by peripheral membrane ruffling and circular dorsal ruffles (CDRs). This further promoted macropinocytosis that mediated the internalization of GAS6-AXL complexes and sustained survival of glioblastoma cells grown under glutamine-deprived conditions. GAS6-induced CDRs contributed to focal adhesion turnover, cell spreading, and elongation. Consequently, AXL activation by GAS6 drove invasion of cancer cells in a spheroid model. All these processes required the kinase activity of AXL, but not TYRO3, and downstream activation of PI3K and RAC1. We propose that GAS6-AXL signaling induces multiple actin-driven cytoskeletal rearrangements that contribute to cancer-cell invasion.
Collapse
Affiliation(s)
- Daria Zdżalik-Bielecka
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland;
| | - Agata Poświata
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Kamila Kozik
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Kamil Jastrzębski
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Kay Oliver Schink
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway
| | | | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Harald Stenmark
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway
| | - Marta Miączyńska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland;
| |
Collapse
|
14
|
Zhou Y, Wang Y, Chen H, Xu Y, Luo Y, Deng Y, Zhang J, Shao A. Immuno-oncology: are TAM receptors in glioblastoma friends or foes? Cell Commun Signal 2021; 19:11. [PMID: 33509214 PMCID: PMC7841914 DOI: 10.1186/s12964-020-00694-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022] Open
Abstract
Tyro3, Axl, and Mertk (TAM) receptors are a subfamily of receptor tyrosine kinases. TAM receptors have been implicated in mediating efferocytosis, regulation of immune cells, secretion of inflammatory factors, and epithelial-to-mesenchymal transition in the tumor microenvironment, thereby serving as a critical player in tumor development and progression. The pro-carcinogenic role of TAM receptors has been widely confirmed, overexpression of TAM receptors is tied to tumor cells growth, metastasis, invasion and treatment resistance. Nonetheless, it is surprising to detect that inhibiting TAM signaling is not all beneficial in the tumor immune microenvironment. The absence of TAM receptors also affects anti-tumor immunity under certain conditions by modulating different immune cells, as the functional diversification of TAM signaling is closely related to tumor immunotherapy. Glioblastoma is the most prevalent and lethal primary brain tumor in adults. Although research regarding the crosstalk between TAM receptors and glioblastoma remains scarce, it appears likely that TAM receptors possess potential anti-tumor effects rather than portraying a total cancer-driving role in the context of glioblastoma. Accordingly, we doubt whether TAM receptors play a double-sided role in glioblastoma, and propose the Janus-faced TAM Hypothesis as a conceptual framework for comprehending the precise underlying mechanisms of TAMs. In this study, we aim to cast a spotlight on the potential multidirectional effects of TAM receptors in glioblastoma and provide a better understanding for TAM receptor-related targeted intervention. Video Abstract
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hailong Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yanyan Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, 211126, Jiangsu, China
| | - Yi Luo
- The Second Affiliated Hospital of Zhejiang University School of Medicine (Changxing Branch), Changxing, Huzhou, 313100, Zhejiang, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
15
|
Wang KH, Ding DC. Dual targeting of TAM receptors Tyro3, Axl, and MerTK: Role in tumors and the tumor immune microenvironment. Tzu Chi Med J 2020; 33:250-256. [PMID: 34386362 PMCID: PMC8323642 DOI: 10.4103/tcmj.tcmj_129_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/12/2020] [Accepted: 07/02/2020] [Indexed: 11/06/2022] Open
Abstract
In both normal and tumor tissues, receptor tyrosine kinases (RTKs) may be pleiotropically expressed. The RTKs not only regulate ordinary cellular processes, including proliferation, survival, adhesion, and migration, but also have a critical role in the development of many types of cancer. The Tyro3, Axl, and MerTK (TAM) family of RTKs (Tyro3, Axl, and MerTK) plays a pleiotropic role in phagocytosis, inflammation, and normal cellular processes. In this article, we highlight the cellular activities of TAM receptors and discuss their roles in cancer and immune cells. We also discuss cancer therapies that target TAM receptors. Further research is needed to elucidate the function of TAM receptors in immune cells toward the development of new targeted immunotherapies for cancer.
Collapse
Affiliation(s)
- Kai-Hung Wang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation and Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
16
|
An induced pluripotent stem cell model of Fanconi anemia reveals mechanisms of p53-driven progenitor cell differentiation. Blood Adv 2020; 4:4679-4692. [PMID: 33002135 DOI: 10.1182/bloodadvances.2020001593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
Fanconi anemia (FA) is a disorder of DNA repair that manifests as bone marrow (BM) failure. The lack of accurate murine models of FA has refocused efforts toward differentiation of patient-derived induced pluripotent stem cells (IPSCs) to hematopoietic progenitor cells (HPCs). However, an intact FA DNA repair pathway is required for efficient IPSC derivation, hindering these efforts. To overcome this barrier, we used inducible complementation of FANCA-deficient IPSCs, which permitted robust maintenance of IPSCs. Modulation of FANCA during directed differentiation to HPCs enabled the production of FANCA-deficient human HPCs that recapitulated FA genotoxicity and hematopoietic phenotypes relative to isogenic FANCA-expressing HPCs. FANCA-deficient human HPCs underwent accelerated terminal differentiation driven by activation of p53/p21. We identified growth arrest specific 6 (GAS6) as a novel target of activated p53 in FANCA-deficient HPCs and modulate GAS6 signaling to rescue hematopoiesis in FANCA-deficient cells. This study validates our strategy to derive a sustainable, highly faithful human model of FA, uncovers a mechanism of HPC exhaustion in FA, and advances toward future cell therapy in FA.
Collapse
|
17
|
Di Stasi R, De Rosa L, D'Andrea LD. Therapeutic aspects of the Axl/Gas6 molecular system. Drug Discov Today 2020; 25:2130-2148. [PMID: 33002607 DOI: 10.1016/j.drudis.2020.09.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/14/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Axl receptor tyrosine kinase (RTK) and its ligand, growth arrest-specific protein 6 (Gas6), are involved in several biological functions and participate in the development and progression of a range of malignancies and autoimmune disorders. In this review, we present this molecular system from a drug discovery perspective, highlighting its therapeutic implications and challenges that need to be addressed. We provide an update on Axl/Gas6 axis biology, exploring its role in fields ranging from angiogenesis, cancer development and metastasis, immune response and inflammation to viral infection. Finally, we summarize the molecules that have been developed to date to target the Axl/Gas6 molecular system for therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Rossella Di Stasi
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Lucia De Rosa
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Luca D D'Andrea
- Istituto di Biostrutture e Bioimmagini, CNR, Via Nizza 52, 10126 Torino, Italy.
| |
Collapse
|
18
|
Broggini T, Piffko A, Hoffmann CJ, Ghori A, Harms C, Adams RH, Vajkoczy P, Czabanka M. Ephrin-B2-EphB4 communication mediates tumor-endothelial cell interactions during hematogenous spread to spinal bone in a melanoma metastasis model. Oncogene 2020; 39:7063-7075. [PMID: 32989254 DOI: 10.1038/s41388-020-01473-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 08/21/2020] [Accepted: 09/15/2020] [Indexed: 11/09/2022]
Abstract
Metastases account for the majority of cancer deaths. Bone represents one of the most common sites of distant metastases, and spinal bone metastasis is the most common source of neurological morbidity in cancer patients. During metastatic seeding of cancer cells, endothelial-tumor cell interactions govern extravasation to the bone and potentially represent one of the first points of action for antimetastatic treatment. The ephrin-B2-EphB4 pathway controls cellular interactions by inducing repulsive or adhesive properties, depending on forward or reverse signaling. Here, we report that in an in vivo metastatic melanoma model, ephrin-B2-mediated activation of EphB4 induces tumor cell repulsion from bone endothelium, translating in reduced spinal bone metastatic loci and improved neurological function. Selective ephrin-B2 depletion in endothelial cells or EphB4 inhibition increases bone metastasis and shortens the time window to hind-limb locomotion deficit from spinal cord compression. EphB4 overexpression in melanoma cells ameliorates the metastatic phenotype and improves neurological outcome. Timely harvesting of bone tissue after tumor cell injection and intravital bone microscopy revealed less tumor cells attached to ephrin-B2-positive endothelial cells. These results suggest that ephrin-B2-EphB4 communication influences bone metastasis formation by altering melanoma cell repulsion/adhesion to bone endothelial cells, and represents a molecular target for therapeutic intervention.
Collapse
Affiliation(s)
- Thomas Broggini
- Department of Neurosurgery, Universitätsmedizin Charite, D-10117, Berlin, Germany.,Department of Physics, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Andras Piffko
- Department of Neurosurgery, Universitätsmedizin Charite, D-10117, Berlin, Germany.,Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Christian J Hoffmann
- Department of Experimental Neurology, Center for Stroke Research Berlin, Universitätsmedizin Charite, D-10117, Berlin, Germany
| | - Adnan Ghori
- Department of Neurosurgery, Universitätsmedizin Charite, D-10117, Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology, Center for Stroke Research Berlin, Universitätsmedizin Charite, D-10117, Berlin, Germany
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Universitätsmedizin Charite, D-10117, Berlin, Germany
| | - Marcus Czabanka
- Department of Neurosurgery, Universitätsmedizin Charite, D-10117, Berlin, Germany.
| |
Collapse
|
19
|
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: 44] [Impact Index Per Article: 11.0] [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
|
20
|
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: 0.3] [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
|
21
|
Rocha-Brito KJP, Fonseca EMB, Oliveira BGDF, Fátima ÂD, Ferreira-Halder CV. Calix[6]arene diminishes receptor tyrosine kinase lifespan in pancreatic cancer cells and inhibits their migration and invasion efficiency. Bioorg Chem 2020; 100:103881. [PMID: 32388429 DOI: 10.1016/j.bioorg.2020.103881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/07/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is a challenging malignancy, mainly due to aggressive regional involvement, early systemic dissemination, high recurrence rate, and subsequent low patient survival. Scientific advances have contributed in particular by identification of molecular targets as well as the definition of the mechanism of action of the drug candidate in the cellular microenvironment. Previously, we have reported the identification of the molecular mechanisms by which calix[6]arene (CLX6) reduces the viability and proliferation of pancreatic cancer cells. Now, we show the biochemical mechanisms by which CLX6 decreases the aggressiveness of Panc-1 cells, focusing specifically on receptor tyrosine kinases (RTK). The results show that clathrin-mediated endocytosis is involved in CLX6-induced AXL receptor tyrosine kinase degradation in Panc-1 cells. This response may be related to the interaction of CLX6 with the tyrosine kinase receptor binding site (such as AXL). As a result, RTK is internalized and degraded by endocytosis, a condition that negatively impacts events dependent on its signaling. Additionally, CLX6 inhibits migration and invasion of Panc-1 cells by downregulating FAK (downstream mediator of AXL) activity and reducing expression levels of MMP2 and MMP9, directly related to the metastatic profile of these cells. It is noteworthy that according to the mechanism proposed here, CLX6 appears as a candidate to be used in therapeutic protocols of patients that display high expression of AXL and consequently, poor diagnosis.
Collapse
Affiliation(s)
- Karin Juliane Pelizzaro Rocha-Brito
- Department of Biochemistry and Tissue Biology, Biology Institute, University of Campinas, Campinas, São Paulo, Brazil; Department of Medicine, Health Sciences Center, University Center of Maringá, Maringá, Paraná, Brazil
| | - Emanuella Maria Barreto Fonseca
- Department of Biochemistry and Tissue Biology, Biology Institute, University of Campinas, Campinas, São Paulo, Brazil; Federal Institute of Education, Science and Technology of São Paulo, São Roque, São Paulo, Brazil
| | | | - Ângelo de Fátima
- Department of Chemistry, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | |
Collapse
|
22
|
Zhou Y, Yao Y, Deng Y, Shao A. Regulation of efferocytosis as a novel cancer therapy. Cell Commun Signal 2020; 18:71. [PMID: 32370748 PMCID: PMC7199874 DOI: 10.1186/s12964-020-00542-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/27/2020] [Indexed: 12/11/2022] Open
Abstract
Efferocytosis is a physiologic phagocytic clearance of apoptotic cells, which modulates inflammatory responses and the immune environment and subsequently facilitates immune escape of cancer cells, thus promoting tumor development and progression. Efferocytosis is an equilibrium formed by perfect coordination among “find-me”, “eat-me” and “don’t-eat-me” signals. These signaling pathways not only affect the proliferation, invasion, metastasis, and angiogenesis of tumor cells but also regulate adaptive responses and drug resistance to antitumor therapies. Therefore, efferocytosis-related molecules and pathways are potential targets for antitumor therapy. Besides, supplementing conventional chemotherapy, radiotherapy and other immunotherapies with efferocytosis-targeted therapy could enhance the therapeutic efficacy, reduce off-target toxicity, and promote patient outcome. Video abstract
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yihan Yao
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
23
|
Meel MH, de Gooijer MC, Metselaar DS, Sewing ACP, Zwaan K, Waranecki P, Breur M, Buil LCM, Lagerweij T, Wedekind LE, Twisk JWR, Koster J, Hashizume R, Raabe EH, Montero Carcaboso Á, Bugiani M, Phoenix TN, van Tellingen O, van Vuurden DG, Kaspers GJL, Hulleman E. Combined Therapy of AXL and HDAC Inhibition Reverses Mesenchymal Transition in Diffuse Intrinsic Pontine Glioma. Clin Cancer Res 2020; 26:3319-3332. [PMID: 32165429 DOI: 10.1158/1078-0432.ccr-19-3538] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/04/2020] [Accepted: 03/06/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Diffuse intrinsic pontine glioma (DIPG) is an incurable type of pediatric brain cancer, which in the majority of cases is driven by mutations in genes encoding histone 3 (H3K27M). We here determined the preclinical therapeutic potential of combined AXL and HDAC inhibition in these tumors to reverse their mesenchymal, therapy-resistant, phenotype. EXPERIMENTAL DESIGN We used public databases and patient-derived DIPG cells to identify putative drivers of the mesenchymal transition in these tumors. Patient-derived neurospheres, xenografts, and allografts were used to determine the therapeutic potential of combined AXL/HDAC inhibition for the treatment of DIPG. RESULTS We identified AXL as a therapeutic target and regulator of the mesenchymal transition in DIPG. Combined AXL and HDAC inhibition had a synergistic and selective antitumor effect on H3K27M DIPG cells. Treatment of DIPG cells with the AXL inhibitor BGB324 and the HDAC inhibitor panobinostat resulted in a decreased expression of mesenchymal and stem cell genes. Moreover, this combination treatment decreased expression of DNA damage repair genes in DIPG cells, strongly sensitizing them to radiation. Pharmacokinetic studies showed that BGB324, like panobinostat, crosses the blood-brain barrier. Consequently, treatment of patient-derived DIPG xenograft and murine DIPG allograft-bearing mice with BGB324 and panobinostat resulted in a synergistic antitumor effect and prolonged survival. CONCLUSIONS Combined inhibition of AXL and HDACs in DIPG cells results in a synergistic antitumor effect by reversing their mesenchymal, stem cell-like, therapy-resistant phenotype. As such, this treatment combination may serve as part of a future multimodal therapeutic strategy for DIPG.
Collapse
Affiliation(s)
- Michaël H Meel
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Mark C de Gooijer
- Division of Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Dennis S Metselaar
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - A Charlotte P Sewing
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Kenn Zwaan
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Piotr Waranecki
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marjolein Breur
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Levi C M Buil
- Division of Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tonny Lagerweij
- Department of Neurosurgery, Neuro-oncology Research Group, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Laurine E Wedekind
- Department of Neurosurgery, Neuro-oncology Research Group, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Jos W R Twisk
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Jan Koster
- Department of Oncogenomics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Rintaro Hashizume
- Departments of Neurological Surgery and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Eric H Raabe
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ángel Montero Carcaboso
- Preclinical Therapeutics and Drug Delivery Research Program, Department of Oncology, Hospital Sant Joan de Déu Barcelona, Spain
| | - Marianna Bugiani
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Timothy N Phoenix
- Division of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati/Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Olaf van Tellingen
- Division of Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Dannis G van Vuurden
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Gertjan J L Kaspers
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Esther Hulleman
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands. .,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
24
|
Ghosh Roy S. TAM receptors: A phosphatidylserine receptor family and its implications in viral infections. TAM RECEPTORS IN HEALTH AND DISEASE 2020; 357:81-122. [DOI: 10.1016/bs.ircmb.2020.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
AXL receptor tyrosine kinase as a promising anti-cancer approach: functions, molecular mechanisms and clinical applications. Mol Cancer 2019; 18:153. [PMID: 31684958 PMCID: PMC6827209 DOI: 10.1186/s12943-019-1090-3] [Citation(s) in RCA: 295] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/18/2019] [Indexed: 02/08/2023] Open
Abstract
Molecular targeted therapy for cancer has been a research hotspot for decades. AXL is a member of the TAM family with the high-affinity ligand growth arrest-specific protein 6 (GAS6). The Gas6/AXL signalling pathway is associated with tumour cell growth, metastasis, invasion, epithelial-mesenchymal transition (EMT), angiogenesis, drug resistance, immune regulation and stem cell maintenance. Different therapeutic agents targeting AXL have been developed, typically including small molecule inhibitors, monoclonal antibodies (mAbs), nucleotide aptamers, soluble receptors, and several natural compounds. In this review, we first provide a comprehensive discussion of the structure, function, regulation, and signalling pathways of AXL. Then, we highlight recent strategies for targeting AXL in the treatment of cancer.AXL-targeted drugs, either as single agents or in combination with conventional chemotherapy or other small molecule inhibitors, are likely to improve the survival of many patients. However, future investigations into AXL molecular signalling networks and robust predictive biomarkers are warranted to select patients who could receive clinical benefit and to avoid potential toxicities.
Collapse
|
26
|
Ellingson BM, Aftab DT, Schwab GM, Hessel C, Harris RJ, Woodworth DC, Leu K, Chakhoyan A, Raymond C, Drappatz J, de Groot J, Prados MD, Reardon DA, Schiff D, Chamberlain M, Mikkelsen T, Desjardins A, Holland J, Ping J, Weitzman R, Wen PY, Cloughesy TF. Volumetric response quantified using T1 subtraction predicts long-term survival benefit from cabozantinib monotherapy in recurrent glioblastoma. Neuro Oncol 2019; 20:1411-1418. [PMID: 29660005 DOI: 10.1093/neuonc/noy054] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background To overcome challenges with traditional response assessment in anti-angiogenic agents, the current study uses T1 subtraction maps to quantify volumetric radiographic response in monotherapy with cabozantinib, an orally bioavailable tyrosine kinase inhibitor with activity against vascular endothelial growth factor receptor 2 (VEGFR2), hepatocyte growth factor receptor (MET), and AXL, in an open-label, phase II trial in patients with recurrent glioblastoma (GBM) (NCT00704288). Methods A total of 108 patients with adequate imaging data and confirmed recurrent GBM were included in this retrospective study from a phase II multicenter trial of cabozantinib monotherapy (XL184-201) at either 100 mg (N = 87) or 140 mg (N = 21) per day. Contrast enhanced T1-weighted digital subtraction maps were used to define volume of contrast-enhancing tumor at baseline and subsequent follow-up time points. Volumetric radiographic response (>65% reduction in contrast-enhancing tumor volume from pretreatment baseline tumor volume sustained for more than 4 wk) was tested as an independent predictor of overall survival (OS). Results Volumetric response rate for all therapeutic doses was 38.9% (41.4% and 28.6% for 100 mg and 140 mg doses, respectively). A log-linear association between baseline tumor volume and OS (P = 0.0006) and a linear correlation between initial change in tumor volume and OS (P = 0.0256) were observed. A significant difference in OS was observed between responders (median OS = 20.6 mo) and nonresponders (median OS = 8.0 mo) (hazard ratio [HR] = 0.3050, P < 0.0001). Multivariable analyses showed that continuous measures of baseline tumor volume (HR = 1.0233, P < 0.0001) and volumetric response (HR = 0.2240, P < 0.0001) were independent predictors of OS. Conclusions T1 subtraction maps provide value in determining response in recurrent GBM treated with cabozantinib and correlated with survival benefit.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | | | | | | | - Robert J Harris
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Davis C Woodworth
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Kevin Leu
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Ararat Chakhoyan
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Jan Drappatz
- Department of Neurology and Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - John de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael D Prados
- Department of Neurosurgery, University of California San Francisco (UCSF), San Francisco, California
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David Schiff
- Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia
| | - Marc Chamberlain
- Department of Neurology, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Annick Desjardins
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | | | - Jerry Ping
- Exelixis, South San Francisco, California
| | | | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
27
|
TAM Receptor Pathways at the Crossroads of Neuroinflammation and Neurodegeneration. DISEASE MARKERS 2019; 2019:2387614. [PMID: 31636733 PMCID: PMC6766163 DOI: 10.1155/2019/2387614] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/04/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023]
Abstract
Increasing evidence suggests that pathogenic mechanisms underlying neurodegeneration are strongly linked with neuroinflammatory responses. Tyro3, Axl, and Mertk (TAM receptors) constitute a subgroup of the receptor tyrosine kinase family, cell surface receptors which transmit signals from the extracellular space to the cytoplasm and nucleus. TAM receptors and the corresponding ligands, Growth Arrest Specific 6 and Protein S, are expressed in different tissues, including the nervous system, playing complex roles in tissue repair, inflammation and cell survival, proliferation, and migration. In the nervous system, TAM receptor signalling modulates neurogenesis and neuronal migration, synaptic plasticity, microglial activation, phagocytosis, myelination, and peripheral nerve repair, resulting in potential interest in neuroinflammatory and neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Multiple Sclerosis. In Alzheimer and Parkinson diseases, a role of TAM receptors in neuronal survival and pathological protein aggregate clearance has been suggested, while in Multiple Sclerosis TAM receptors are involved in myelination and demyelination processes. To better clarify roles and pathways involving TAM receptors may have important therapeutic implications, given the fine modulation of multiple molecular processes which could be reached. In this review, we summarise the roles of TAM receptors in the central nervous system, focusing on the regulation of immune responses and microglial activities and analysing in vitro and in vivo studies regarding TAM signalling involvement in neurodegeneration.
Collapse
|
28
|
Sun ZG, Liu JH, Zhang JM, Qian Y. Research Progress of Axl Inhibitors. Curr Top Med Chem 2019; 19:1338-1349. [PMID: 31218961 DOI: 10.2174/1568026619666190620155613] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/24/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023]
Abstract
Axl, a Receptor Tyrosine Kinase (RTK) belonging to the TAM (Axl, Mer, Tyro3) family, participates in many signal transduction cascades after mostly being stimulated by Growth arrestspecific 6(Gas6). Axl is widely expressed in many organs, such as macrophages, endothelial cells, heart, liver and skeletal muscle. Over-expression and activation of Axl are associated with promoting chemotherapy resistance, cell proliferation, invasion and metastasis in many human cancers, such as breast, lung, and pancreatic cancers. Therefore, the research and development of Axl inhibitors is of great significance to strengthen the means of cancer treatment, especially to solve the problem of drug resistance. Axl inhibitors have attracted more and more researchers' attention in recent years. This review discusses the research progress of Axl inhibitors in recent years.
Collapse
Affiliation(s)
- Zhi-Gang Sun
- Central Laboratory, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400, China.,State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing 210023, China
| | - Jian-Hua Liu
- Central Laboratory, Linyi Central Hospital, No.17 Jiankang Road, Linyi 276400, China
| | - Jin-Mai Zhang
- Room 205, BIO-X white house, Shanghai Jiao Tong University, No.1954 Huashan Road, Shanghai 200030, China
| | - Yong Qian
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing 210023, China
| |
Collapse
|
29
|
Yan J, Xu C, Li Y, Tang B, Xie S, Hong T, Zeng E. Long non-coding RNA LINC00526 represses glioma progression via forming a double negative feedback loop with AXL. J Cell Mol Med 2019; 23:5518-5531. [PMID: 31240814 PMCID: PMC6653591 DOI: 10.1111/jcmm.14435] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/04/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022] Open
Abstract
Glioma is the most common primary intracranial carcinoma with extremely poor prognosis. The significances of long non‐coding RNA (lncRNA) involved in glioma have been started revealed. However, the expression, roles and molecular mechanisms of most lncRNAs in glioma are still unknown. In this study, we identified a novel lncRNA LINC00526, which is significantly low expressed in glioma. Low expression of LINC00526 is correlated with aggravation and poor survival in glioma. Functional assays revealed that ectopic expression of LINC00526 inhibits glioma cell proliferation, migration, and invasion. LINC00526 silencing promotes glioma cell proliferation, migration and invasion. Mechanistically, we found that LINC00526 directly interacts with EZH2, represses the binding of EZH2 to AXL promoter, attenuates the transcriptional activating roles of EZH2 on AXL, and therefore represses AXL expression. Via repressing AXL, LINC00526 further represses PI3K/Akt/NF‐κB signalling. Intriguingly, we identified that NFKB1 and NFKB2 directly binds LINC00526 promoter and represses LINC00526 transcription. We further found that via activating NF‐κB signalling, AXL represses LINC00526 transcription. Therefore, LINC00526/EZH2/AXL/PI3K/Akt/NF‐κB form a feedback loop in glioma. Analysis of the TCGA data revealed that the expression of LINC00526 is inversely correlated with that of AXL in glioma tissues. In addition, functional rescue assays revealed that the tumour suppressive roles of LINC00526 are dependent on the negative regulation of AXL. Collectively, our data identified LINC00526 as a tumour suppressor in glioma via forming a double negative feedback loop with AXL. Our data also suggested LINC00526 as a potential prognostic biomarker and therapeutic candidate for glioma.
Collapse
Affiliation(s)
- Jian Yan
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chunhua Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Youping Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Tang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shenhao Xie
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Erming Zeng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
30
|
Zhang Y, Sui R, Chen Y, Liang H, Shi J, Piao H. Downregulation of miR-485-3p promotes glioblastoma cell proliferation and migration via targeting RNF135. Exp Ther Med 2019; 18:475-482. [PMID: 31258684 PMCID: PMC6566029 DOI: 10.3892/etm.2019.7600] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 04/11/2019] [Indexed: 01/04/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that serve pivotal roles in human diseases. Several miRNAs, such as miR-485-3p, have been identified as potential biomarkers for predicting overall survival of patients with glioblastoma (GBM). However, the underlying mechanism of miRNAs in promoting GBM progression remains unknown. In the present study, decreased miR-485-3p expression was detected in tumor tissues from patients with GBM. Using western blot analysis, reverse transcription-quantitative PCR and dual luciferase reporter assay, ring finger protein 135 (RNF135) was confirmed as a target gene of miR-485-3p in GBM cells. Through silencing of RNF135, miR-485-3p inactivated the mitogen-activated protein kinase/ERK1/2 pathway in GBM cells. Moreover, functional assays demonstrated that miR-485-3p inhibited GBM cell proliferation and migration whilst overexpression of RNF135 reversed this effect. Additionally, a negative correlation between miR-485-3p and RNF135 mRNA expression was observed in tissues from patients with glioblastoma. In conclusion, the present results demonstrated that miR-485-3p functioned as a tumor suppressor which suggested that miR-485-3p might have a role in GBM progression.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Rui Sui
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Yi Chen
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Haiyang Liang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Ji Shi
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
31
|
Cloughesy TF, Drappatz J, de Groot J, Prados MD, Reardon DA, Schiff D, Chamberlain M, Mikkelsen T, Desjardins A, Ping J, Holland J, Weitzman R, Wen PY. Phase II study of cabozantinib in patients with progressive glioblastoma: subset analysis of patients with prior antiangiogenic therapy. Neuro Oncol 2019; 20:259-267. [PMID: 29036345 PMCID: PMC5777491 DOI: 10.1093/neuonc/nox151] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Cabozantinib is a potent, multitarget inhibitor of MET and vascular endothelial growth factor receptor 2 (VEGFR2). This open-label, phase II trial evaluated cabozantinib in patients with recurrent or progressive glioblastoma (GBM). Methods Patients were initially enrolled to a starting cabozantinib dose of 140 mg/day, but the starting dose was amended to 100 mg/day because of safety concerns. Treatment continued until disease progression or unacceptable toxicity. The primary endpoint was objective response rate, assessed by an independent radiology facility using modified Response Assessment in Neuro-Oncology criteria. Additional endpoints included duration of response, 6-month and median progression-free survival, overall survival, glucocorticoid use, and safety. Results Among 222 patients enrolled, 70 had received prior antiangiogenic therapy. Herein, we report results in this subset of 70 patients. The objective response rate was 4.3%, and the median duration of response was 4.2 months. The proportion of patients alive and progression free at 6 months was 8.5%. Median progression-free survival was 2.3 months, and median overall survival was 4.6 months. The most common adverse events reported in all patients, regardless of dose group, included fatigue (74.3%), diarrhea (47.1%), increased alanine aminotransferase (37.1%), headache (35.7%), hypertension (35.7%), and nausea (35.7%); overall, 34 (48.6%) patients experienced adverse events that resulted in dose reductions. Conclusions Cabozantinib treatment appeared to have modest clinical activity with a 4.3% response rate in patients who had received prior antiangiogenic therapy for GBM. Clinical Trials Registration Number NCT00704288 (https://www.clinicaltrials.gov/ct2/show/NCT00704288)
Collapse
Affiliation(s)
- Timothy F Cloughesy
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Jan Drappatz
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - John de Groot
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Michael D Prados
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - David A Reardon
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - David Schiff
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Marc Chamberlain
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Tom Mikkelsen
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Annick Desjardins
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Jerry Ping
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Jaymes Holland
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Ron Weitzman
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Patrick Y Wen
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| |
Collapse
|
32
|
Wen PY, Drappatz J, de Groot J, Prados MD, Reardon DA, Schiff D, Chamberlain M, Mikkelsen T, Desjardins A, Holland J, Ping J, Weitzman R, Cloughesy TF. Phase II study of cabozantinib in patients with progressive glioblastoma: subset analysis of patients naive to antiangiogenic therapy. Neuro Oncol 2019; 20:249-258. [PMID: 29016998 PMCID: PMC5777496 DOI: 10.1093/neuonc/nox154] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Cabozantinib is a tyrosine kinase inhibitor with activity against vascular endothelial growth factor receptor 2 (VEGFR2) and MET that has demonstrated clinical activity in advanced solid tumors. This open-label, phase II trial evaluated cabozantinib in patients with recurrent or refractory glioblastoma (GBM). Methods Patients were initially enrolled at a starting dose of 140 mg/day, but the starting dose was amended to 100 mg/day because of toxicity. Treatment continued until disease progression or unacceptable toxicity. The primary endpoint was objective response rate assessed by an independent radiology facility using modified Response Assessment in Neuro-Oncology criteria. Additional endpoints included duration of response, 6-month and median progression-free survival, overall survival, and safety. Results Among 152 patients naive to prior antiangiogenic therapy, the objective response rate was 17.6% and 14.5% in the 140 mg/day and 100 mg/day groups, respectively, which did not meet the predefined statistical target for success. The proportions of patients alive and progression free at 6 months were 22.3% and 27.8%, respectively. Median progression-free survival was 3.7 months in both groups, and median overall survival was 7.7 months and 10.4 months, respectively. The incidence of grade 3/4 adverse events (AEs) was 79.4% and 84.7% in the 140 mg/day and 100 mg/day groups, respectively, and dose reductions due to AEs were experienced by 61.8% and 72.0%, respectively. Common grade 3/4 AEs included fatigue, diarrhea, and palmar-plantar erythrodysesthesia syndrome. Conclusions Cabozantinib showed evidence of clinical activity in patients with recurrent GBM naive to antiangiogenic therapy, although the predefined statistical target for success was not met. At the starting doses assessed, AEs were frequently managed with dose reductions. Clinical Trials Registration Number NCT00704288 (https://www.clinicaltrials.gov/ct2/show/NCT00704288).
Collapse
Affiliation(s)
- Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Jan Drappatz
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - John de Groot
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Michael D Prados
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - David Schiff
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Marc Chamberlain
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Tom Mikkelsen
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Annick Desjardins
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Jaymes Holland
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Jerry Ping
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Ron Weitzman
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Timothy F Cloughesy
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| |
Collapse
|
33
|
Baird AM, Easty D, Jarzabek M, Shiels L, Soltermann A, Klebe S, Raeppel S, MacDonagh L, Wu C, Griggs K, Kirschner MB, Stanfill B, Nonaka D, Goparaju CM, Murer B, Fennell DA, O'Donnell DM, Barr MP, Mutti L, Reid G, Finn S, Cuffe S, Pass HI, Opitz I, Byrne AT, O'Byrne KJ, Gray SG. When RON MET TAM in Mesothelioma: All Druggable for One, and One Drug for All? Front Endocrinol (Lausanne) 2019; 10:89. [PMID: 30863365 PMCID: PMC6399142 DOI: 10.3389/fendo.2019.00089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive inflammatory cancer with a poor survival rate. Treatment options are limited at best and drug resistance is common. Thus, there is an urgent need to identify novel therapeutic targets in this disease in order to improve patient outcomes and survival times. MST1R (RON) is a trans-membrane receptor tyrosine kinase (RTK), which is part of the c-MET proto-oncogene family. The only ligand recognized to bind MST1R (RON) is Macrophage Stimulating 1 (MST1), also known as Macrophage Stimulating Protein (MSP) or Hepatocyte Growth Factor-Like Protein (HGFL). In this study, we demonstrate that the MST1-MST1R (RON) signaling axis is active in MPM. Targeting this pathway with a small molecule inhibitor, LCRF-0004, resulted in decreased proliferation with a concomitant increase in apoptosis. Cell cycle progression was also affected. Recombinant MST1 treatment was unable to overcome the effect of LCRF-0004 in terms of either proliferation or apoptosis. Subsequently, the effect of an additional small molecular inhibitor, BMS-777607 (which targets MST1R (RON), MET, Tyro3, and Axl) also resulted in a decreased proliferative capacity of MPM cells. In a cohort of MPM patient samples, high positivity for total MST1R by IHC was an independent predictor of favorable prognosis. Additionally, elevated expression levels of MST1 also correlated with better survival. This study also determined the efficacy of LCRF-0004 and BMS-777607 in xenograft MPM models. Both LCRF-0004 and BMS-777607 demonstrated significant anti-tumor efficacy in vitro, however BMS-777607 was far superior to LCRF-0004. The in vivo and in vitro data generated by this study indicates that a multi-TKI, targeting the MST1R/MET/TAM signaling pathways, may provide a more effective therapeutic strategy for the treatment of MPM as opposed to targeting MST1R alone.
Collapse
Affiliation(s)
- Anne-Marie Baird
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
- Cancer and Ageing Research Program, Queensland University of Technology, Brisbane, QLD, Australia
| | - David Easty
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
| | - Monika Jarzabek
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Liam Shiels
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alex Soltermann
- Department of Clinical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Sonja Klebe
- Department of Anatomical Pathology, Flinders University of South Australia, Bedford Park, SA, Australia
| | | | - Lauren MacDonagh
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
| | - Chengguang Wu
- Department of Clinical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Kim Griggs
- Department of Anatomical Pathology, Flinders University of South Australia, Bedford Park, SA, Australia
| | - Michaela B. Kirschner
- Asbestos Diseases Research Institute, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, NSW, Australia
| | - Bryan Stanfill
- The Commonwealth Scientific and Industrial Research Organization, Brisbane, QLD, Australia
| | - Daisuke Nonaka
- Department of Histopathology, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Department of Cardiothoracic Surgery, New York University (NYU) Langone Medical Center, New York, NY, United States
| | - Chandra M. Goparaju
- Department of Cardiothoracic Surgery, New York University (NYU) Langone Medical Center, New York, NY, United States
| | - Bruno Murer
- Department of Clinical Pathology, Ospedale dell'Angelo, Venice, Italy
| | - Dean A. Fennell
- MRC Toxicology Unit, University of Leicester and Leicester University Hospitals, Leicester, United Kingdom
| | | | - Martin P. Barr
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
| | - Luciano Mutti
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Glen Reid
- Asbestos Diseases Research Institute, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, NSW, Australia
| | - Stephen Finn
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin, Ireland
| | - Sinead Cuffe
- HOPE Directorate, St James's Hospital, Dublin, Ireland
| | - Harvey I. Pass
- Department of Cardiothoracic Surgery, New York University (NYU) Langone Medical Center, New York, NY, United States
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Annette T. Byrne
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kenneth J. O'Byrne
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
- Cancer and Ageing Research Program, Queensland University of Technology, Brisbane, QLD, Australia
- HOPE Directorate, St James's Hospital, Dublin, Ireland
- Division of Cancer Services, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Steven G. Gray
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
- *Correspondence: Steven G. Gray
| |
Collapse
|
34
|
The Dual Role of TAM Receptors in Autoimmune Diseases and Cancer: An Overview. Cells 2018; 7:cells7100166. [PMID: 30322068 PMCID: PMC6210017 DOI: 10.3390/cells7100166] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/05/2018] [Accepted: 10/09/2018] [Indexed: 01/01/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) regulate cellular processes by converting signals from the extracellular environment to the cytoplasm and nucleus. Tyro3, Axl, and Mer (TAM) receptors form an RTK family that plays an intricate role in tissue maintenance, phagocytosis, and inflammation as well as cell proliferation, survival, migration, and development. Defects in TAM signaling are associated with numerous autoimmune diseases and different types of cancers. Here, we review the structure of TAM receptors, their ligands, and their biological functions. We discuss the role of TAM receptors and soluble circulating TAM receptors in the autoimmune diseases systemic lupus erythematosus (SLE) and multiple sclerosis (MS). Lastly, we discuss the effect of TAM receptor deregulation in cancer and explore the therapeutic potential of TAM receptors in the treatment of diseases.
Collapse
|
35
|
Shafit-Zagardo B, Gruber RC, DuBois JC. The role of TAM family receptors and ligands in the nervous system: From development to pathobiology. Pharmacol Ther 2018. [PMID: 29514053 DOI: 10.1016/j.pharmthera.2018.03.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tyro3, Axl, and Mertk, referred to as the TAM family of receptor tyrosine kinases, are instrumental in maintaining cell survival and homeostasis in mammals. TAM receptors interact with multiple signaling molecules to regulate cell migration, survival, phagocytosis and clearance of metabolic products and cell debris called efferocytosis. The TAMs also function as rheostats to reduce the expression of proinflammatory molecules and prevent autoimmunity. All three TAM receptors are activated in a concentration-dependent manner by the vitamin K-dependent growth arrest-specific protein 6 (Gas6). Gas6 and the TAMs are abundantly expressed in the nervous system. Gas6, secreted by neurons and endothelial cells, is the sole ligand for Axl. ProteinS1 (ProS1), another vitamin K-dependent protein functions mainly as an anti-coagulant, and independent of this function can activate Tyro3 and Mertk, but not Axl. This review will focus on the role of the TAM receptors and their ligands in the nervous system. We highlight studies that explore the function of TAM signaling in myelination, the visual cortex, neural cancers, and multiple sclerosis (MS) using Gas6-/- and TAM mutant mice models.
Collapse
Affiliation(s)
- Bridget Shafit-Zagardo
- Albert Einstein College of Medicine, Department of Pathology, 1300 Morris Park Avenue, Bronx, NY 10461, United States.
| | - Ross C Gruber
- Sanofi, Neuroinflammation and MS Research, 49 New York Ave, Framingham, MA 01701, United States
| | - Juwen C DuBois
- Albert Einstein College of Medicine, Department of Pathology, 1300 Morris Park Avenue, Bronx, NY 10461, United States
| |
Collapse
|
36
|
Antony J, Huang RYJ. AXL-Driven EMT State as a Targetable Conduit in Cancer. Cancer Res 2017; 77:3725-3732. [PMID: 28667075 DOI: 10.1158/0008-5472.can-17-0392] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/19/2017] [Accepted: 05/22/2017] [Indexed: 12/14/2022]
Abstract
The receptor tyrosine kinase (RTK) AXL has been intrinsically linked to epithelial-mesenchymal transition (EMT) and promoting cell survival, anoikis resistance, invasion, and metastasis in several cancers. AXL signaling has been shown to directly affect the mesenchymal state and confer it with aggressive phenotype and drug resistance. Recently, the EMT gradient has also been shown to rewire the kinase signaling nodes that facilitate AXL-RTK cross-talk, protracted signaling, converging on ERK, and PI3K axes. The molecular mechanisms underplaying the regulation between the kinome and EMT require further elucidation to define targetable conduits. Therapeutically, as AXL inhibition has shown EMT reversal and resensitization to other tyrosine kinase inhibitors, mitotic inhibitors, and platinum-based therapy, there is a need to stratify patients based on AXL dependence. This review elucidates the role of AXL in EMT-mediated oncogenesis and highlights the reciprocal control between AXL signaling and the EMT state. In addition, we review the potential in inhibiting AXL for the development of different therapeutic strategies and inhibitors. Cancer Res; 77(14); 3725-32. ©2017 AACR.
Collapse
Affiliation(s)
- Jane Antony
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.,Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Ruby Yun-Ju Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore. .,Department of Obstetrics and Gynecology, National University Health System, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
37
|
Onken J, Vajkoczy P, Torka R, Hempt C, Patsouris V, Heppner FL, Radke J. Phospho-AXL is widely expressed in glioblastoma and associated with significant shorter overall survival. Oncotarget 2017; 8:50403-50414. [PMID: 28881571 PMCID: PMC5584143 DOI: 10.18632/oncotarget.18468] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/10/2017] [Indexed: 11/25/2022] Open
Abstract
Receptor tyrosine kinase AXL (RTK-AXL) is regarded as a suitable target in glioblastoma (GBM) therapy. Since AXL kinase inhibitors are about to get approval for clinical use, patients with a potential benefit from therapy targeting AXL need to be identified. We therefore assessed the expression pattern of Phospho-AXL (P-AXL), the biologically active form of AXL, in 90 patients with newly diagnosed GBM, which was found to be detectable in 67 patients (corresponding to 74%). We identified three main P-AXL expression patterns: i) exclusively in the tumor vasculature (13%), ii) in areas of hypercellularity (35%), or iii) both, in the tumor vasculature and in hypercellular areas of the tumor tissue (52%). Pattern iii) is associated with significant decrease in overall survival (Hazard ratio 2.349, 95% confidence interval 1.069 to 5.162, *p=0.03). Our data suggest that P-AXL may serve as a therapeutic target in the majority of GBM patients.
Collapse
Affiliation(s)
- Julia Onken
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Torka
- Institute of Physiological Chemistry, Martin Luther University of Halle-Wittenberg, Halle/Saale, Germany
| | - Claudia Hempt
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Victor Patsouris
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank L Heppner
- Berlin Institute of Health (BIH), Berlin, Germany.,Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Cluster of Excellence, NeuroCure, Berlin, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany, Partner Site Charité Berlin, Berlin, Germany
| | - Josefine Radke
- Berlin Institute of Health (BIH), Berlin, Germany.,Department of Neuropathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany, Partner Site Charité Berlin, Berlin, Germany
| |
Collapse
|
38
|
n-Butylidenephthalide Regulated Tumor Stem Cell Genes EZH2/AXL and Reduced Its Migration and Invasion in Glioblastoma. Int J Mol Sci 2017; 18:ijms18020372. [PMID: 28208648 PMCID: PMC5343907 DOI: 10.3390/ijms18020372] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/06/2017] [Indexed: 01/16/2023] Open
Abstract
Glioblastoma (GBM) is one of the most common and aggressive types of brain tumor. Due to its highly recurrent rate and poor prognosis, the overall survival time with this type of tumor is only 20–21 months. Recent knowledge suggests that its recurrence is in part due to the presence of cancer stem cells (CSCs), which display radioresistant, chemoresistant, self-renewal and tumorigenic potential. Enhancers of Zeste 2 (EZH2) and AXL receptor tyrosine kinase (AXL) are both highly expressed in GBM. Additionally, they are an essential regulator involved in CSCs maintenance, migration, invasion, epithelial-to-mesenchymal transition (EMT), stemness, metastasis and patient survival. In this study, we used a small molecule, n-butylidenephthalide (BP), to assess the anti-GBM stem-like cells potential, and then tried to find out the associated genes involved with regulation in migration and invasion. We demonstrated that BP reduced the expression of AXL and stemness related genes in a dose-dependent manner. The migratory and invasive capabilities of GBM stem-like cells could be reduced by AXL/EZH2. Finally, in the overexpression of AXL, EZH2 and Sox2 by transfection in GBM stem-like cells, we found that AXL/EZH2/TGF-β1, but not Sox2, might be a key regulator in tumor invasion, migration and EMT. These results might help in the development of a new anticancer compound and can be a target for treating GBM.
Collapse
|
39
|
Choi SW, Lee KS, Lee JH, Kang HJ, Lee MJ, Kim HY, Park KI, Kim SL, Shin HK, Seo WD. Suppression of Akt-HIF-1α signaling axis by diacetyl atractylodiol inhibits hypoxia-induced angiogenesis. BMB Rep 2017; 49:508-13. [PMID: 27439603 PMCID: PMC5227144 DOI: 10.5483/bmbrep.2016.49.9.069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Indexed: 12/16/2022] Open
Abstract
Hypoxia-inducible factor (HIF)-1α is a key regulator associated with tumorigenesis, angiogenesis, and metastasis. HIF-1α regulation under hypoxia has been highlighted as a promising therapeutic target in angiogenesis-related diseases. Here, we demonstrate that diacetyl atractylodiol (DAA) from Atractylodes japonica (A. japonica) is a potent HIF-1α inhibitor that inhibits the Akt signaling pathway. DAA dose-dependently inhibited hypoxia-induced HIF-1α and downregulated Akt signaling without affecting the stability of HIF-1α protein. Furthermore, DAA prevented hypoxia-mediated angiogenesis based on in vitro tube formation and in vivo chorioallantoic membrane (CAM) assays. Therefore, DAA might be useful for treatment of hypoxia-related tumorigenesis, including angiogenesis. [BMB Reports 2016; 49(9): 508-513]
Collapse
Affiliation(s)
- Sik-Won Choi
- Division of Crop Foundation, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365, Korea
| | - Kwang-Sik Lee
- Division of Crop Foundation, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365; College of Crop Science and Biotechnology, Dankook University, Cheonan 31116, Korea
| | - Jin Hwan Lee
- Division of Research Development and Education, National Institute of Chemical Safety, Ministry of Environment, Daejeon 34111, Korea
| | - Hyeon Jung Kang
- Division of Crop Foundation, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365, Korea
| | - Mi Ja Lee
- Division of Crop Foundation, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365, Korea
| | - Hyun Young Kim
- Division of Crop Foundation, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365, Korea
| | - Kie-In Park
- Division of Biological Sciences, College of Natural Science, Chonbuk National University, Jeonju 54896, Korea
| | - Sun-Lim Kim
- Division of Crop Foundation, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365, Korea
| | - Hye Kyoung Shin
- Department of surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| | - Woo Duck Seo
- Division of Crop Foundation, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365, Korea
| |
Collapse
|
40
|
Gay CM, Balaji K, Byers LA. Giving AXL the axe: targeting AXL in human malignancy. Br J Cancer 2017; 116:415-423. [PMID: 28072762 PMCID: PMC5318970 DOI: 10.1038/bjc.2016.428] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 12/14/2022] Open
Abstract
The receptor tyrosine kinase AXL, activated by a complex interaction between its ligand growth arrest-specific protein 6 and phosphatidylserine, regulates various vital cellular processes, including proliferation, survival, motility, and immunologic response. Although not implicated as an oncogenic driver itself, AXL, a member of the TYRO3, AXL, and MERTK family of receptor tyrosine kinases, is overexpressed in several haematologic and solid malignancies, including acute myeloid leukaemia, non-small cell lung cancer, gastric and colorectal adenocarcinomas, and breast and prostate cancers. In the context of malignancy, evidence suggests that AXL overexpression drives wide-ranging processes, including epithelial to mesenchymal transition, tumour angiogenesis, resistance to chemotherapeutic and targeted agents, and decreased antitumor immune response. As a result, AXL is an attractive candidate not only as a prognostic biomarker in malignancy but also as a target for anticancer therapies. Several AXL inhibitors are currently in preclinical and clinical development. This article reviews the structure, regulation, and function of AXL; the role of AXL in the tumour microenvironment; the development of AXL as a therapeutic target; and areas of ongoing and future investigation.
Collapse
Affiliation(s)
- Carl M Gay
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Kavitha Balaji
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Lauren Averett Byers
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| |
Collapse
|
41
|
Duma CM, Kim BS, Chen PV, Plunkett ME, Mackintosh R, Mathews MS, Casserly RM, Mendez GA, Furman DJ, Smith G, Oh N, Caraway CA, Sanathara AR, Dillman RO, Riley AS, Weiland D, Stemler L, Cannell R, Abrams DA, Smith A, Owen CM, Eisenberg B, Brant-Zawadzki M. Upfront boost Gamma Knife “leading-edge” radiosurgery to FLAIR MRI–defined tumor migration pathways in 174 patients with glioblastoma multiforme: a 15-year assessment of a novel therapy. J Neurosurg 2016; 125:40-49. [DOI: 10.3171/2016.7.gks161460] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVEGlioblastoma multiforme (GBM) is composed of cells that migrate through the brain along predictable white matter pathways. Targeting white matter pathways adjacent to, and leading away from, the original contrast-enhancing tumor site (termed leading-edge radiosurgery [LERS]) with single-fraction stereotactic radiosurgery as a boost to standard therapy could limit the spread of glioma cells and improve clinical outcomes.METHODSBetween December 2000 and May 2016, after an initial diagnosis of GBM and prior to or during standard radiation therapy and carmustine or temozolomide chemotherapy, 174 patients treated with radiosurgery to the leading edge (LE) of tumor cell migration were reviewed. The LE was defined as a region outside the contrast-enhancing tumor nidus, defined by FLAIR MRI. The median age of patients was 59 years (range 22–87 years). Patients underwent LERS a median of 18 days from original diagnosis. The median target volume of 48.5 cm3 (range 2.5–220.0 cm3) of LE tissue was targeted using a median dose of 8 Gy (range 6–14 Gy) at the 50% isodose line.RESULTSThe median overall survival was 23 months (mean 43 months) from diagnosis. The 2-, 3-, 5-, 7-, and 10-year actual overall survival rates after LERS were 39%, 26%, 16%, 10%, and 4%, respectively. Nine percent of patients developed treatment-related imaging-documented changes due to LERS. Nineteen percent of patients were hospitalized for management of edema, 22% for resection of a tumor cyst or new tumor bulk, and 2% for shunting to treat hydrocephalus throughout the course of their disease. Of the patients still alive, Karnofsky Performance Scale scores remained stable in 90% of patients and decreased by 1–3 grades in 10% due to symptomatic treatment-related imaging changes.CONCLUSIONSLERS is a safe and effective upfront adjunctive therapy for patients with newly diagnosed GBM. Limitations of this study include a single-center experience and single-institution determination of the LE tumor target. Use of a leading-edge calculation algorithm will be described to achieve a consistent approach to defining the LE target for general use. A multicenter trial will further elucidate its value in the treatment of GBM.
Collapse
Affiliation(s)
| | - Brian S. Kim
- 2Cancer Center, and
- 3Department of Radiation Oncology, Hoag Memorial Hospital Presbyterian, Newport Beach
| | - Peter V. Chen
- 2Cancer Center, and
- 3Department of Radiation Oncology, Hoag Memorial Hospital Presbyterian, Newport Beach
| | - Marianne E. Plunkett
- 2Cancer Center, and
- 3Department of Radiation Oncology, Hoag Memorial Hospital Presbyterian, Newport Beach
| | - Ralph Mackintosh
- 2Cancer Center, and
- 3Department of Radiation Oncology, Hoag Memorial Hospital Presbyterian, Newport Beach
| | - Marlon S. Mathews
- 4Department of Neurosurgery, University of California, Irvine, Orange; and
| | | | | | | | | | - Nathan Oh
- 1Neurosciences Institute,
- 5Department of Neurosurgery, Loma Linda University Health, Loma Linda, California
| | | | | | | | | | | | | | | | | | - Alexa Smith
- 4Department of Neurosurgery, University of California, Irvine, Orange; and
| | | | | | | |
Collapse
|
42
|
Rinschen MM, Schroeter CB, Koehler S, Ising C, Schermer B, Kann M, Benzing T, Brinkkoetter PT. Quantitative deep mapping of the cultured podocyte proteome uncovers shifts in proteostatic mechanisms during differentiation. Am J Physiol Cell Physiol 2016; 311:C404-17. [PMID: 27357545 DOI: 10.1152/ajpcell.00121.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 06/27/2016] [Indexed: 11/22/2022]
Abstract
The renal filtration barrier is maintained by the renal podocyte, an epithelial postmitotic cell. Immortalized mouse podocyte cell lines-both in the differentiated and undifferentiated state-are widely utilized tools to estimate podocyte injury and cytoskeletal rearrangement processes in vitro. Here, we mapped the cultured podocyte proteome at a depth of more than 8,800 proteins and quantified 7,240 proteins. Copy numbers of proteins mutated in forms of hereditary nephrotic syndrome or focal segmental glomerulosclerosis (FSGS) were assessed. We found that cultured podocytes express abundant copy numbers of endogenous receptors, such as tyrosine kinase membrane receptors, the G protein-coupled receptor (GPCR), NPR3 (ANP receptor), and several poorly characterized GPCRs. The data set was correlated with deep mapping mRNA sequencing ("mRNAseq") data from the native mouse podocyte, the native mouse podocyte proteome and staining intensities from the human protein atlas. The generated data set was similar to these previously published resources, but several native and high-abundant podocyte-specific proteins were not identified in the data set. Notably, this data set detected general perturbations in proteostatic mechanisms as a dominant alteration during podocyte differentiation, with high proteasome activity in the undifferentiated state and markedly increased expression of lysosomal proteins in the differentiated state. Phosphoproteomics analysis of mouse podocytes at a resolution of more than 3,000 sites suggested a preference of phosphorylation of actin filament-associated proteins in the differentiated state. The data set obtained here provides a resource and provides the means for deep mapping of the native podocyte proteome and phosphoproteome in a similar manner.
Collapse
Affiliation(s)
- Markus M Rinschen
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany; and Systems Biology of Ageing Cologne, SybaCol, Cologne, Germany
| | - Christina B Schroeter
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Sybille Koehler
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Christina Ising
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany; and Systems Biology of Ageing Cologne, SybaCol, Cologne, Germany
| | - Martin Kann
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany; and Systems Biology of Ageing Cologne, SybaCol, Cologne, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
43
|
Chang C, Ortiz K, Ansari A, Gershwin ME. The Zika outbreak of the 21st century. J Autoimmun 2016; 68:1-13. [PMID: 26925496 PMCID: PMC7127657 DOI: 10.1016/j.jaut.2016.02.006] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 12/25/2022]
Abstract
The Zika virus outbreak has captivated the attention of the global audience and information has spread rapidly and wildly through the internet and other media channels. This virus was first identified in 1947, when it was isolated from a sentinel rhesus monkey placed by British scientists working at the Yellow Fever Research Laboratory located in the Zika forest area of Uganda, hence its name, and is transmitted primarily by the mosquito vector, Aedes aegypti. The fact that the rhesus macaque is an Asian species being placed in an African forest brings to mind the possibility of rapid adaptation of the virus from an African to Asian species, an issue that has not been considered. Whether such adaptation has played any role in acquiring pathogenicity due to cross species transmission remains to be identified. The first human infection was described in Nigeria in 1954, with only scattered reports of about a dozen human infections identified over a 50-year period. It was not until 2007 that Zika virus raised its ugly head with infections noted in three-quarters of the population on the tiny island of Yap located between the Philippines and Papua New Guinea in the western Pacific Ocean, followed by a major outbreak in French Polynesia in 2013. The virus remained confined to a narrow equatorial band in Africa and Asia until 2014 when it began to spread eastward, first toward Oceania and then to South America. Since then, millions of infected individuals have been identified in Brazil, Colombia, Venezuela, including 25 additional countries in the Americas. While the symptoms associated with Zika virus infection are generally mild, consisting of fever, maculopapular rash, arthralgia and conjunctivitis, there have been reports of more severe reactions that are associated with neurological complications. In pregnant women, fetal neurological complications include brain damage and microcephaly, while in adults there have been several cases of virus-associated Guillain-Barre syndrome. The virus was until recently believed to only be transmitted via mosquitoes. But when the Zika virus was isolated from the semen specimens from a patient in Texas, this provided the basis for the recent report of possible sexual transmission of the Zika virus. Due to the neurological complications, various vectors for infection as well as the rapid spread throughout the globe, it has prompted the World Health Organization to issue a global health emergency. Various governmental organizations have recommended that pregnant women do not travel to countries where the virus is epidemic, and within the countries affected by the virus, recommendations were provided for women of childbearing age to delay pregnancy. The overall public health impact of these above findings highlights the need for a rapid but specific diagnostic test for blood banks worldwide to identify those infected and for the counseling of women who are pregnant or contemplating pregnancy. As of this date, there are neither commercially licensed diagnostic tests nor a vaccine. Because cross-reactivity of the Zika virus with dengue and Chikungunya virus is common, it may pose difficulty in being able to quickly develop such tests and vaccines. So far the most effective public health measures include controlling the mosquito populations via insecticides and preventing humans from direct exposure to mosquitoes.
Collapse
Affiliation(s)
- Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, Davis, CA, USA
| | - Kristina Ortiz
- Departmenty of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Aftab Ansari
- Departmenty of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, Davis, CA, USA.
| |
Collapse
|