1
|
Tomuleasa C, Tigu AB, Munteanu R, Moldovan CS, Kegyes D, Onaciu A, Gulei D, Ghiaur G, Einsele H, Croce CM. Therapeutic advances of targeting receptor tyrosine kinases in cancer. Signal Transduct Target Ther 2024; 9:201. [PMID: 39138146 PMCID: PMC11323831 DOI: 10.1038/s41392-024-01899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 08/15/2024] Open
Abstract
Receptor tyrosine kinases (RTKs), a category of transmembrane receptors, have gained significant clinical attention in oncology due to their central role in cancer pathogenesis. Genetic alterations, including mutations, amplifications, and overexpression of certain RTKs, are critical in creating environments conducive to tumor development. Following their discovery, extensive research has revealed how RTK dysregulation contributes to oncogenesis, with many cancer subtypes showing dependency on aberrant RTK signaling for their proliferation, survival and progression. These findings paved the way for targeted therapies that aim to inhibit crucial biological pathways in cancer. As a result, RTKs have emerged as primary targets in anticancer therapeutic development. Over the past two decades, this has led to the synthesis and clinical validation of numerous small molecule tyrosine kinase inhibitors (TKIs), now effectively utilized in treating various cancer types. In this manuscript we aim to provide a comprehensive understanding of the RTKs in the context of cancer. We explored the various alterations and overexpression of specific receptors across different malignancies, with special attention dedicated to the examination of current RTK inhibitors, highlighting their role as potential targeted therapies. By integrating the latest research findings and clinical evidence, we seek to elucidate the pivotal role of RTKs in cancer biology and the therapeutic efficacy of RTK inhibition with promising treatment outcomes.
Collapse
Affiliation(s)
- Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania.
| | - Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Cristian-Silviu Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - David Kegyes
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Anca Onaciu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hermann Einsele
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Universitätsklinikum Würzburg, Medizinische Klinik II, Würzburg, Germany
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Su D, Ding C, Qiu J, Yang G, Wang R, Liu Y, Tao J, Luo W, Weng G, Zhang T. Ribosome profiling: a powerful tool in oncological research. Biomark Res 2024; 12:11. [PMID: 38273337 PMCID: PMC10809610 DOI: 10.1186/s40364-024-00562-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/12/2024] [Indexed: 01/27/2024] Open
Abstract
Neoplastic cells need to adapt their gene expression pattern to survive in an ever-changing or unfavorable tumor microenvironment. Protein synthesis (or mRNA translation), an essential part of gene expression, is dysregulated in cancer. The emergence of distinct translatomic technologies has revolutionized oncological studies to elucidate translational regulatory mechanisms. Ribosome profiling can provide adequate information on diverse aspects of translation by aiding in quantitatively analyzing the intensity of translating ribosome-protected fragments. Here, we review the primary currently used translatomics techniques and highlight their advantages and disadvantages as tools for translatomics studies. Subsequently, we clarified the areas in which ribosome profiling could be applied to better understand translational control. Finally, we summarized the latest advances in cancer studies using ribosome profiling to highlight the extensive application of this powerful and promising translatomic tool.
Collapse
Affiliation(s)
- Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Chen Ding
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Gang Yang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Ruobing Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Wenhao Luo
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Guihu Weng
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China.
| |
Collapse
|
3
|
Jones VT, Graves-Deal R, Cao Z, Bogatcheva G, Ramirez MA, Harmych SJ, Higginbotham JN, Sharma V, Damalanka VC, Wahoski CC, Joshi N, Irudayam MJ, Roland JT, Ayers GD, Liu Q, Coffey RJ, Janetka JW, Singh B. Inhibition of autocrine HGF maturation overcomes cetuximab resistance in colorectal cancer. Cell Mol Life Sci 2024; 81:28. [PMID: 38212428 PMCID: PMC10784391 DOI: 10.1007/s00018-023-05071-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/27/2023] [Accepted: 11/27/2023] [Indexed: 01/13/2024]
Abstract
Although amplifications and mutations in receptor tyrosine kinases (RTKs) act as bona fide oncogenes, in most cancers, RTKs maintain moderate expression and remain wild-type. Consequently, cognate ligands control many facets of tumorigenesis, including resistance to anti-RTK therapies. Herein, we show that the ligands for the RTKs MET and RON, HGF and HGFL, respectively, are synthesized as inactive precursors that are activated by cellular proteases. Our newly generated HGF/HGFL protease inhibitors could overcome both de novo and acquired cetuximab resistance in colorectal cancer (CRC). Conversely, HGF overexpression was necessary and sufficient to induce cetuximab resistance and loss of polarity. Moreover, HGF-induced cetuximab resistance could be overcome by the downstream MET inhibitor, crizotinib, and upstream protease inhibitors. Additionally, HAI-1, an endogenous inhibitor of HGF proteases, (i) was downregulated in CRC, (ii) exhibited increased genomic methylation that correlated with poor prognosis, (iii) HAI-1 expression correlated with cetuximab response in a panel of cancer cell lines, and (iv) exogenous addition of recombinant HAI-1 overcame cetuximab resistance in CC-HGF cells. Thus, we describe a targetable, autocrine HAI-1/Protease/HGF/MET axis in cetuximab resistance in CRC.
Collapse
Affiliation(s)
- Vivian Truong Jones
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ramona Graves-Deal
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Zheng Cao
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Galina Bogatcheva
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Marisol A Ramirez
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sarah J Harmych
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - James N Higginbotham
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Vineeta Sharma
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Vishnu C Damalanka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Claudia C Wahoski
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Neeraj Joshi
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Maria Johnson Irudayam
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
| | - Joseph T Roland
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Gregory D Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - James W Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Bhuminder Singh
- Department of Medicine, Vanderbilt University Medical Center, 10465J, MRB IV, 2213 Garland Avenue, Nashville, TN, 37232-0441, USA.
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
4
|
Shikalov A, Koman I, Kogan NM. Targeted Glioma Therapy-Clinical Trials and Future Directions. Pharmaceutics 2024; 16:100. [PMID: 38258110 PMCID: PMC10820492 DOI: 10.3390/pharmaceutics16010100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of glioma, with a median survival of 14.6 months post-diagnosis. Understanding the molecular profile of such tumors allowed the development of specific targeted therapies toward GBM, with a major role attributed to tyrosine kinase receptor inhibitors and immune checkpoint inhibitors. Targeted therapeutics are drugs that work by specific binding to GBM-specific or overexpressed markers on the tumor cellular surface and therefore contain a recognition moiety linked to a cytotoxic agent, which produces an antiproliferative effect. In this review, we have summarized the available information on the targeted therapeutics used in clinical trials of GBM and summarized current obstacles and advances in targeted therapy concerning specific targets present in GBM tumor cells, outlined efficacy endpoints for major classes of investigational drugs, and discussed promising strategies towards an increase in drug efficacy in GBM.
Collapse
Affiliation(s)
| | | | - Natalya M. Kogan
- Department of Molecular Biology, Institute of Personalized and Translational Medicine, Ariel University, Ariel 40700, Israel; (A.S.); (I.K.)
| |
Collapse
|
5
|
Ramar V, Guo S, Hudson B, Liu M. Progress in Glioma Stem Cell Research. Cancers (Basel) 2023; 16:102. [PMID: 38201528 PMCID: PMC10778204 DOI: 10.3390/cancers16010102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Glioblastoma multiforme (GBM) represents a diverse spectrum of primary tumors notorious for their resistance to established therapeutic modalities. Despite aggressive interventions like surgery, radiation, and chemotherapy, these tumors, due to factors such as the blood-brain barrier, tumor heterogeneity, glioma stem cells (GSCs), drug efflux pumps, and DNA damage repair mechanisms, persist beyond complete isolation, resulting in dismal outcomes for glioma patients. Presently, the standard initial approach comprises surgical excision followed by concurrent chemotherapy, where temozolomide (TMZ) serves as the foremost option in managing GBM patients. Subsequent adjuvant chemotherapy follows this regimen. Emerging therapeutic approaches encompass immunotherapy, including checkpoint inhibitors, and targeted treatments, such as bevacizumab, aiming to exploit vulnerabilities within GBM cells. Nevertheless, there exists a pressing imperative to devise innovative strategies for both diagnosing and treating GBM. This review emphasizes the current knowledge of GSC biology, molecular mechanisms, and associations with various signals and/or pathways, such as the epidermal growth factor receptor, PI3K/AKT/mTOR, HGFR/c-MET, NF-κB, Wnt, Notch, and STAT3 pathways. Metabolic reprogramming in GSCs has also been reported with the prominent activation of the glycolytic pathway, comprising aldehyde dehydrogenase family genes. We also discuss potential therapeutic approaches to GSC targets and currently used inhibitors, as well as their mode of action on GSC targets.
Collapse
Affiliation(s)
- Vanajothi Ramar
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (V.R.); (B.H.)
| | - Shanchun Guo
- Department of Chemistry, Xavier University, 1 Drexel Dr., New Orleans, LA 70125, USA;
| | - BreAnna Hudson
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (V.R.); (B.H.)
| | - Mingli Liu
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (V.R.); (B.H.)
| |
Collapse
|
6
|
Qin A, Qin Y, Lee J, Musket A, Ying M, Krenciute G, Marincola FM, Yao ZQ, Musich PR, Xie Q. Tyrosine kinase signaling-independent MET-targeting with CAR-T cells. J Transl Med 2023; 21:682. [PMID: 37779207 PMCID: PMC10544186 DOI: 10.1186/s12967-023-04521-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/11/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND Recent progress in cancer immunotherapy encourages the expansion of chimeric antigen receptor (CAR) T cell therapy in solid tumors including hepatocellular carcinoma (HCC). Overexpression of MET receptor tyrosine kinase is common in HCC; however, MET inhibitors are effective only when MET is in an active form, making patient stratification difficult. Specific MET-targeting CAR-T cells hold the promise of targeting HCC with MET overexpression regardless of signaling pathway activity. METHODS MET-specific CARs with CD28ζ or 4-1BBζ as co-stimulation domains were constructed. MET-CAR-T cells derived from healthy subjects (HS) and HCC patients were evaluated for their killing activity and cytokine release against HCC cells with various MET activations in vitro, and for their tumor growth inhibition in orthotopic xenograft models in vivo. RESULTS MET-CAR.CD28ζ and MET-CAR.4-1BBζ T cells derived from both HS and HCC patients specifically killed MET-positive HCC cells. When stimulated with MET-positive HCC cells in vitro, MET-CAR.CD28ζ T cells demonstrated a higher level of cytokine release and expression of programmed cell death protein 1 (PD-1) than MET-CAR.4-1BBζ T cells. When analyzed in vivo, MET-CAR.CD28ζ T cells more effectively inhibited HCC orthotopic tumor growth in mice when compared to MET-CAR.4-1BBζ T cells. CONCLUSION We generated and characterized MET-specific CAR-T cells for targeting HCC with MET overexpression regardless of MET activation. Compared with MET-CAR.4-1BBζ, MET-CAR.CD28ζ T cells showed a higher anti-HCC potency but also a higher level of T cell exhaustion. While MET-CAR.CD28ζ is preferred for further development, overcoming the exhaustion of MET-CAR-T cells is necessary to improve their therapeutic efficacy in vivo.
Collapse
Affiliation(s)
- Anna Qin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Yuan Qin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Joseph Lee
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Anna Musket
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Mingyao Ying
- Department of Neurology, Hugo W. Moser Research Institute at Kennedy Krieger, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | | | - Zhi Q Yao
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Phillip R Musich
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Qian Xie
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
| |
Collapse
|
7
|
Krajcer A, Grzywna E, Lewandowska-Łańcucka J. Strategies increasing the effectiveness of temozolomide at various levels of anti-GBL therapy. Biomed Pharmacother 2023; 165:115174. [PMID: 37459661 DOI: 10.1016/j.biopha.2023.115174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/17/2023] Open
Abstract
Glioblastoma (GBL) is the most common (60-70% of primary brain tumours) and the most malignant of the glial tumours. Although current therapies remain palliative, they have been proven to prolong overall survival. Within an optimal treatment regimen (incl. surgical resection, radiation therapy, and chemotherapy) temozolomide as the current anti-GBL first-line chemotherapeutic has increased the median overall survival to 14-15 months, and the percentage of patients alive at two years has been reported to rise from 10.4% to 26.5%. Though, the effectiveness of temozolomide chemotherapy is limited by the serious systemic, dose-related side effects. Therefore, the ponderation regarding novel treatment methods along with innovative formulations is crucial to emerging the therapeutic potential of the widely used drug simultaneously reducing the drawbacks of its use. Herein the complex temozolomide application restrictions present at different levels of therapy as well as, the currently proposed strategies aimed at reducing those limitations are demonstrated. Approaches increasing the efficacy of anti-GBL treatment are addressed. Our paper is focused on the most recent developments in the field of nano/biomaterials-based systems for temozolomide delivery and their functionalization towards more effective blood-brain-barrier crossing and/or tumour targeting. Appropriate designing accounting for the physical and chemical features of formulations along with distinct routes of administration is also discussed. In addition, considering the multiple resistance mechanisms, the molecular heterogeneity and the evolution of tumour the purposely selected delivery methods, the combined therapeutic approaches and specifically focused on GBL cells therapies are reviewed.
Collapse
Affiliation(s)
- Aleksandra Krajcer
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Ewelina Grzywna
- Department of Neurosurgery and Neurotraumatology, Jagiellonian University Medical College, Św. Anny 12, 31-008 Kraków, Poland
| | | |
Collapse
|
8
|
Zhong J, Wu X, Gao Y, Chen J, Zhang M, Zhou H, Yang J, Xiao F, Yang X, Huang N, Qi H, Wang X, Bai F, Shi Y, Zhang N. Circular RNA encoded MET variant promotes glioblastoma tumorigenesis. Nat Commun 2023; 14:4467. [PMID: 37491377 PMCID: PMC10368723 DOI: 10.1038/s41467-023-40212-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/18/2023] [Indexed: 07/27/2023] Open
Abstract
Activated by its single ligand, hepatocyte growth factor (HGF), the receptor tyrosine kinase MET is pivotal in promoting glioblastoma (GBM) stem cell self-renewal, invasiveness and tumorigenicity. Nevertheless, HGF/MET-targeted therapy has shown limited clinical benefits in GBM patients, suggesting hidden mechanisms of MET signalling in GBM. Here, we show that circular MET RNA (circMET) encodes a 404-amino-acid MET variant (MET404) facilitated by the N6-methyladenosine (m6A) reader YTHDF2. Genetic ablation of circMET inhibits MET404 expression in mice and attenuates MET signalling. Conversely, MET404 knock-in (KI) plus P53 knock-out (KO) in mouse astrocytes initiates GBM tumorigenesis and shortens the overall survival. MET404 directly interacts with the MET β subunit and forms a constitutively activated MET receptor whose activity does not require HGF stimulation. High MET404 expression predicts poor prognosis in GBM patients, indicating its clinical relevance. Targeting MET404 through a neutralizing antibody or genetic ablation reduces GBM tumorigenicity in vitro and in vivo, and combinatorial benefits are obtained with the addition of a traditional MET inhibitor. Overall, we identify a MET variant that promotes GBM tumorigenicity, offering a potential therapeutic strategy for GBM patients, especially those with MET hyperactivation.
Collapse
Affiliation(s)
- Jian Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China
| | - Xujia Wu
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China
| | - Yixin Gao
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China
| | - Junju Chen
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China
| | - Maolei Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China
| | - Huangkai Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China
| | - Jia Yang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China
| | - Feizhe Xiao
- Department of Scientific Research Section, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xuesong Yang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China
| | - Nunu Huang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China
| | - Haoyue Qi
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Laboratory of Tumour Immunopathology of the Ministry of Education of China, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiuxing Wang
- National Health Commission Key Laboratory of Antibody Techniques, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University (PKU), Beijing, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China.
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Key Laboratory of Tumour Immunopathology of the Ministry of Education of China, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
9
|
Singh H. Role of Molecular Targeted Therapeutic Drugs in Treatment of Glioblastoma: A Review Article. Glob Med Genet 2023; 10:42-47. [PMID: 37077370 PMCID: PMC10110362 DOI: 10.1055/s-0043-57028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023] Open
Abstract
Glioblastoma is remarkably periodic primary brain tumor, characterizing an eminently heterogeneous pattern of neoplasms that are utmost destructive and threatening cancers. An enhanced and upgraded knowledge of the various molecular pathways that cause malignant changes in glioblastoma has resulted in advancement of numerous biomarkers and the interpretation of various agents that pointedly target tumor cells and microenvironment. In this review, literature or information on various targeted therapy for glioblastoma is discussed. English language articles were scrutinized in plentiful directory or databases like PubMed, ScienceDirect, Web of Sciences, Google Scholar, and Scopus. The important keywords used for searching databases are "Glioblastoma," "Targeted therapy in glioblastoma," "Therapeutic drugs in glioblastoma," and "Molecular targets in glioblastoma."
Collapse
Affiliation(s)
- Himanshu Singh
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, Index Institute of Dental Sciences, Indore, Madhya Pradesh, India
| |
Collapse
|
10
|
Saunders JT, Kumar S, Benavides-Serrato A, Holmes B, Benavides KE, Bashir MT, Nishimura RN, Gera J. Translation of circHGF RNA encodes an HGF protein variant promoting glioblastoma growth through stimulation of c-MET. J Neurooncol 2023; 163:207-218. [PMID: 37162666 DOI: 10.1007/s11060-023-04331-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/28/2023] [Indexed: 05/11/2023]
Abstract
INTRODUCTION HGF/c-MET signaling is a significant driver of glioblastoma (GBM) growth and disease progression. Unfortunately, c-MET targeted therapies have been found to be largely ineffective suggesting additional redundant mechanisms of c-MET activation. METHODS Utilizing RNA-sequencing (RNA-seq) and ribosome profiling analyses of circular RNAs, circ-HGF (hsa_circ_0080914) was identified as markedly upregulated in primary GBM and found to potentially encode an HGF protein variant (C-HGF) 119 amino acids in length. This candidate HGF variant was characterized and evaluated for its ability to mediate c-MET activation and regulate PDX GBM cell growth, motility and invasive potential in vitro and tumor burden in intracranial xenografts in mice. RESULTS An internal ribosome entry site (IRES) was identified within the circ-HGF RNA which mediated translation of the cross-junctional ORF encoding C-HGF and was observed to be highly expressed in GBM relative to normal brain tissue. C-HGF was also found to be secreted from GBM cells and concentrated cell culture supernatants or recombinant C-HGF activated known signaling cascades downstream of c-MET. C-HGF was shown to interact directly with the c-MET receptor resulting in its autophosphorylation and activation in PDX GBM lines. Knockdown of C-HGF resulted in suppression of c-MET signaling and marked inhibition of cell growth, motility and invasiveness, whereas overexpression of C-HGF displayed the opposite effects. Additionally, modulation of C-HGF expression regulated tumor growth in intracranial xenografted PDX GBM models. CONCLUSIONS These results reveal an alternative mechanism of c-MET activation via a circular RNA encoded HGF protein variant which is relevant in GBM biology. Targeting C-HGF may offer a promising approach for GBM clinical management.
Collapse
Affiliation(s)
- Jacquelyn T Saunders
- Department of Medicine, David Geffen School of Medicine at UCLA, University of California-Los Angeles, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA
| | - Sunil Kumar
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA
| | - Angelica Benavides-Serrato
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA
| | - Brent Holmes
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA
| | - Kennedy E Benavides
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA
| | - Muhammad T Bashir
- Department of Medicine, David Geffen School of Medicine at UCLA, University of California-Los Angeles, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA
| | - Robert N Nishimura
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California-Los Angeles, Los Angeles, USA
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA
| | - Joseph Gera
- Department of Medicine, David Geffen School of Medicine at UCLA, University of California-Los Angeles, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA.
- Jonnson Comprehensive Cancer Center, University of California-Los Angeles, Greater Los Angeles Veterans Affairs Healthcare System, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA.
- Molecular Biology Institute, University of California-Los Angeles, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA.
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, 16111 Plummer Street (151), Building 1, Room C111A, Los Angeles, CA, 91343, USA.
| |
Collapse
|
11
|
Warashina S, Sato H, Zouda M, Takahashi M, Wada Y, Passioura T, Suga H, Watanabe Y, Matsumoto K, Mukai H. Two-Chain Mature Hepatocyte Growth Factor-Specific Positron Emission Tomography Imaging in Tumors Using 64Cu-Labeled HiP-8, a Nonstandard Macrocyclic Peptide Probe. Mol Pharm 2023; 20:2029-2038. [PMID: 36862642 DOI: 10.1021/acs.molpharmaceut.2c01020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Two-chain hepatocyte growth factor (tcHGF), the mature form of HGF, is associated with malignancy and anticancer drug resistance; therefore, its quantification is an important indicator for cancer diagnosis. In tumors, activated tcHGF hardly discharges into the systemic circulation, indicating that tcHGF is an excellent target for molecular imaging using positron emission tomography (PET). We recently discovered HGF-inhibitory peptide-8 (HiP-8) that binds specifically to human tcHGF with nanomolar affinity. The purpose of this study was to investigate the usefulness of HiP-8-based PET probes in human HGF knock-in humanized mice. 64Cu-labeled HiP-8 molecules were synthesized using a cross-bridged cyclam chelator, CB-TE1K1P. Radio-high-performance liquid chromatography-based metabolic stability analyses showed that more than 90% of the probes existed in intact form in blood at least for 15 min. In PET studies, significantly selective visualization of hHGF-overexpressing tumors versus hHGF-negative tumors was observed in double-tumor-bearing mice. The accumulation of labeled HiP-8 into the hHGF-overexpressing tumors was significantly reduced by competitive inhibition. In addition, the radioactivity and distribution of phosphorylated MET/HGF receptor were colocalized in tissues. These results demonstrate that the 64Cu-labeled HiP-8 probes are suitable for tcHGF imaging in vivo, and secretory proteins like tcHGF can be a target for PET imaging.
Collapse
Affiliation(s)
- Shota Warashina
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hiroki Sato
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Maki Zouda
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Maiko Takahashi
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuhiro Wada
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Toby Passioura
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo 113-0033, Japan
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo 113-0033, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan.,WPI-Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma, Kanazawa, Ishikawa 920-1192, Japan
| | - Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,Department of Pharmaceutical Informatics, Graduate School of Biomedical Science, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
12
|
Cecchi F, Rex K, Schmidt J, Vocke CD, Lee YH, Burkett S, Baker D, Damore MA, Coxon A, Burgess TL, Bottaro DP. Rilotumumab Resistance Acquired by Intracrine Hepatocyte Growth Factor Signaling. Cancers (Basel) 2023; 15:460. [PMID: 36672409 PMCID: PMC9857108 DOI: 10.3390/cancers15020460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Drug resistance is a long-standing impediment to effective systemic cancer therapy and acquired drug resistance is a growing problem for molecularly-targeted therapeutics that otherwise have shown unprecedented successes in disease control. The hepatocyte growth factor (HGF)/Met receptor pathway signaling is frequently involved in cancer and has been a subject of targeted drug development for nearly 30 years. To anticipate and study specific resistance mechanisms associated with targeting this pathway, we engineered resistance to the HGF-neutralizing antibody rilotumumab in glioblastoma cells harboring autocrine HGF/Met signaling, a frequent abnormality of this brain cancer in humans. We found that rilotumumab resistance was acquired through an unusual mechanism comprising dramatic HGF overproduction and misfolding, endoplasmic reticulum (ER) stress-response signaling and redirected vesicular trafficking that effectively sequestered rilotumumab and misfolded HGF from native HGF and activated Met. Amplification of MET and HGF genes, with evidence of rapidly acquired intron-less, reverse-transcribed copies in DNA, was also observed. These changes enabled persistent Met pathway activation and improved cell survival under stress conditions. Point mutations in the HGF pathway or other complementary or downstream growth regulatory cascades that are frequently associated with targeted drug resistance in other prevalent cancer types were not observed. Although resistant cells were significantly more malignant, they retained sensitivity to Met kinase inhibition and acquired sensitivity to inhibition of ER stress signaling and cholesterol biosynthesis. Defining this mechanism reveals details of a rapidly acquired yet highly-orchestrated multisystem route of resistance to a selective molecularly-targeted agent and suggests strategies for early detection and effective intervention.
Collapse
Affiliation(s)
- Fabiola Cecchi
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karen Rex
- Amgen, Inc., Thousand Oaks, CA 91320, USA
| | | | - Cathy D. Vocke
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Young H. Lee
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sandra Burkett
- Molecular Cytogenetics Core Facility, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | | | | | | | | | - Donald P. Bottaro
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
Li S, Wang C, Chen J, Lan Y, Zhang W, Kang Z, Zheng Y, Zhang R, Yu J, Li W. Signaling pathways in brain tumors and therapeutic interventions. Signal Transduct Target Ther 2023; 8:8. [PMID: 36596785 PMCID: PMC9810702 DOI: 10.1038/s41392-022-01260-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 01/05/2023] Open
Abstract
Brain tumors, although rare, contribute to distinct mortality and morbidity at all ages. Although there are few therapeutic options for brain tumors, enhanced biological understanding and unexampled innovations in targeted therapies and immunotherapies have considerably improved patients' prognoses. Nonetheless, the reduced response rates and unavoidable drug resistance of currently available treatment approaches have become a barrier to further improvement in brain tumor (glioma, meningioma, CNS germ cell tumors, and CNS lymphoma) treatment. Previous literature data revealed that several different signaling pathways are dysregulated in brain tumor. Importantly, a better understanding of targeting signaling pathways that influences malignant behavior of brain tumor cells might open the way for the development of novel targeted therapies. Thus, there is an urgent need for a more comprehensive understanding of the pathogenesis of these brain tumors, which might result in greater progress in therapeutic approaches. This paper began with a brief description of the epidemiology, incidence, risk factors, as well as survival of brain tumors. Next, the major signaling pathways underlying these brain tumors' pathogenesis and current progress in therapies, including clinical trials, targeted therapies, immunotherapies, and system therapies, have been systemically reviewed and discussed. Finally, future perspective and challenges of development of novel therapeutic strategies in brain tumor were emphasized.
Collapse
Affiliation(s)
- Shenglan Li
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Can Wang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinyi Chen
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanjie Lan
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weichunbai Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhuang Kang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yi Zheng
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Rong Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jianyu Yu
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
14
|
Small Molecule Inhibitors for Hepatocellular Carcinoma: Advances and Challenges. Molecules 2022; 27:molecules27175537. [PMID: 36080304 PMCID: PMC9457820 DOI: 10.3390/molecules27175537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 12/12/2022] Open
Abstract
According to data provided by World Health Organization, hepatocellular carcinoma (HCC) is the sixth most common cause of deaths due to cancer worldwide. Tremendous progress has been achieved over the last 10 years developing novel agents for HCC treatment, including small-molecule kinase inhibitors. Several small molecule inhibitors currently form the core of HCC treatment due to their versatility since they would be more easily absorbed and have higher oral bioavailability, thus easier to formulate and administer to patients. In addition, they can be altered structurally to have greater volumes of distribution, allowing them to block extravascular molecular targets and to accumulate in a high concentration in the tumor microenvironment. Moreover, they can be designed to have shortened half-lives to control for immune-related adverse events. Most importantly, they would spare patients, healthcare institutions, and society as a whole from the burden of high drug costs. The present review provides an overview of the pharmaceutical compounds that are licensed for HCC treatment and other emerging compounds that are still investigated in preclinical and clinical trials. These molecules are targeting different molecular targets and pathways that are proven to be involved in the pathogenesis of the disease.
Collapse
|
15
|
Montella L, Del Gaudio N, Bove G, Cuomo M, Buonaiuto M, Costabile D, Visconti R, Facchini G, Altucci L, Chiariotti L, Della Monica R. Looking Beyond the Glioblastoma Mask: Is Genomics the Right Path? Front Oncol 2022; 12:926967. [PMID: 35875139 PMCID: PMC9306486 DOI: 10.3389/fonc.2022.926967] [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: 04/23/2022] [Accepted: 06/09/2022] [Indexed: 11/15/2022] Open
Abstract
Glioblastomas are the most frequent and malignant brain tumor hallmarked by an invariably poor prognosis. They have been classically differentiated into primary isocitrate dehydrogenase 1 or 2 (IDH1 -2) wild-type (wt) glioblastoma (GBM) and secondary IDH mutant GBM, with IDH wt GBMs being commonly associated with older age and poor prognosis. Recently, genetic analyses have been integrated with epigenetic investigations, strongly implementing typing and subtyping of brain tumors, including GBMs, and leading to the new WHO 2021 classification. GBM genomic and epigenomic profile influences evolution, resistance, and therapeutic responses. However, differently from other tumors, there is a wide gap between the refined GBM profiling and the limited therapeutic opportunities. In addition, the different oncogenes and tumor suppressor genes involved in glial cell transformation, the heterogeneous nature of cancer, and the restricted access of drugs due to the blood–brain barrier have limited clinical advancements. This review will summarize the more relevant genetic alterations found in GBMs and highlight their potential role as potential therapeutic targets.
Collapse
Affiliation(s)
- Liliana Montella
- Oncology Operative Unit, "Santa Maria delle Grazie" Hospital, ASL Napoli 2 NORD-, Pozzuoli, Italy
| | - Nunzio Del Gaudio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Guglielmo Bove
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Mariella Cuomo
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| | - Michela Buonaiuto
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| | - Davide Costabile
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,SEMM-European School of Molecular Medicine, Milano, Italy
| | - Roberta Visconti
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Institute of Experimental Endocrinology and Oncology, Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Gaetano Facchini
- Oncology Operative Unit, "Santa Maria delle Grazie" Hospital, ASL Napoli 2 NORD-, Pozzuoli, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy.,BIOGEM, Ariano Irpino, Italy
| | - Lorenzo Chiariotti
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| | - Rosa Della Monica
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| |
Collapse
|
16
|
Therapeutic Strategies for Ovarian Cancer in Point of HGF/c-MET Targeting. Medicina (B Aires) 2022; 58:medicina58050649. [PMID: 35630066 PMCID: PMC9147666 DOI: 10.3390/medicina58050649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/09/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022] Open
Abstract
Ovarian cancer is the fifth leading cause of cancer deaths in women and is regarded as one of the most difficult cancers to treat. Currently, studies are being conducted to develop therapeutic agents for effective treatment of ovarian cancer. In this review, we explain the properties of the hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (c-MET) and how the signaling pathway of HGF/c-MET is activated in different cancers and involved in tumorigenesis and metastasis of ovarian cancer. We present the findings of clinical studies using small chemicals or antibodies targeting HGF/c-MET signaling in various cancer types, particularly in ovarian cancer. We also discuss that HGF/c-MET-targeted therapy, when combined with chemo drugs, could be an effective strategy for ovarian cancer therapeutics.
Collapse
|
17
|
Immunotherapeutic Approaches for Glioblastoma Treatment. Biomedicines 2022; 10:biomedicines10020427. [PMID: 35203636 PMCID: PMC8962267 DOI: 10.3390/biomedicines10020427] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma remains a challenging disease to treat, despite well-established standard-of-care treatments, with a median survival consistently of less than 2 years. In this review, we delineate the unique disease-specific challenges for immunotherapies, both brain-related and non-brain-related, which will need to be adequately overcome for the development of effective treatments. We also review current immunotherapy treatments, with a focus on clinical applications, and propose future directions for the field of GBM immunotherapy.
Collapse
|
18
|
Yang K, Wu Z, Zhang H, Zhang N, Wu W, Wang Z, Dai Z, Zhang X, Zhang L, Peng Y, Ye W, Zeng W, Liu Z, Cheng Q. Glioma targeted therapy: insight into future of molecular approaches. Mol Cancer 2022; 21:39. [PMID: 35135556 PMCID: PMC8822752 DOI: 10.1186/s12943-022-01513-z] [Citation(s) in RCA: 331] [Impact Index Per Article: 165.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
Gliomas are the common type of brain tumors originating from glial cells. Epidemiologically, gliomas occur among all ages, more often seen in adults, which males are more susceptible than females. According to the fifth edition of the WHO Classification of Tumors of the Central Nervous System (WHO CNS5), standard of care and prognosis of gliomas can be dramatically different. Generally, circumscribed gliomas are usually benign and recommended to early complete resection, with chemotherapy if necessary. Diffuse gliomas and other high-grade gliomas according to their molecule subtype are slightly intractable, with necessity of chemotherapy. However, for glioblastoma, feasible resection followed by radiotherapy plus temozolomide chemotherapy define the current standard of care. Here, we discuss novel feasible or potential targets for treatment of gliomas, especially IDH-wild type glioblastoma. Classic targets such as the p53 and retinoblastoma (RB) pathway and epidermal growth factor receptor (EGFR) gene alteration have met failure due to complex regulatory network. There is ever-increasing interest in immunotherapy (immune checkpoint molecule, tumor associated macrophage, dendritic cell vaccine, CAR-T), tumor microenvironment, and combination of several efficacious methods. With many targeted therapy options emerging, biomarkers guiding the prescription of a particular targeted therapy are also attractive. More pre-clinical and clinical trials are urgently needed to explore and evaluate the feasibility of targeted therapy with the corresponding biomarkers for effective personalized treatment options.
Collapse
Affiliation(s)
- Keyang Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijing Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Nan Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,One-Third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wantao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xun Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yun Peng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China.,Teaching and Research Section of Clinical Nursing, Xiangya Hospital of Central South University, Changsha, China
| | - Weijie Ye
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenjing Zeng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
19
|
Qin A, Musket A, Musich PR, Schweitzer JB, Xie Q. Receptor tyrosine kinases as druggable targets in glioblastoma: Do signaling pathways matter? Neurooncol Adv 2021; 3:vdab133. [PMID: 34806012 PMCID: PMC8598918 DOI: 10.1093/noajnl/vdab133] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant primary brain tumor without effective therapies. Since bevacizumab was FDA approved for targeting vascular endothelial growth factor receptor 2 (VEGFR2) in adult patients with recurrent GBM, targeted therapy against receptor tyrosine kinases (RTKs) has become a new avenue for GBM therapeutics. In addition to VEGFR, the epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGFR), hepatocyte growth factor receptor (HGFR/MET), and fibroblast growth factor receptor (FGFR) are major RTK targets. However, results from clinical Phase II/III trials indicate that most RTK-targeting therapeutics including tyrosine kinase inhibitors (TKIs) and neutralizing antibodies lack clinical efficacy, either alone or in combination. The major challenge is to uncover the genetic RTK alterations driving GBM initiation and progression, as well as to elucidate the mechanisms toward therapeutic resistance. In this review, we will discuss the genetic alterations in these 5 commonly targeted RTKs, the clinical trial outcomes of the associated RTK-targeting therapeutics, and the potential mechanisms toward the resistance. We anticipate that future design of new clinical trials with combination strategies, based on the genetic alterations within an individual patient’s tumor and mechanisms contributing to therapeutic resistance after treatment, will achieve durable remissions and improve outcomes in GBM patients.
Collapse
Affiliation(s)
- Anna Qin
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Anna Musket
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Phillip R Musich
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - John B Schweitzer
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Qian Xie
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| |
Collapse
|
20
|
Fu J, Su X, Li Z, Deng L, Liu X, Feng X, Peng J. HGF/c-MET pathway in cancer: from molecular characterization to clinical evidence. Oncogene 2021; 40:4625-4651. [PMID: 34145400 DOI: 10.1038/s41388-021-01863-w] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/17/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023]
Abstract
This review provides a comprehensive landscape of HGF/c-MET (hepatocyte growth factor (HGF) /mesenchymal-epithelial transition factor (c-MET)) signaling pathway in cancers. First, we generalize the compelling influence of HGF/c-MET pathway on multiple cellular processes. Then, we present the genomic characterization of HGF/c-MET pathway in carcinogenesis. Furthermore, we extensively illustrate the malignant biological behaviors of HGF/c-MET pathway in cancers, in which hyperactive HGF/c-MET signaling is considered as a hallmark. In addition, we investigate the current clinical trials of HGF/c-MET-targeted therapy in cancers. We find that although HGF/c-MET-targeted therapy has led to breakthroughs in certain cancers, monotherapy of targeting HGF/c-MET has failed to demonstrate significant clinical efficacy in most cancers. With the advantage of the combinations of HGF/c-MET-targeted therapy, the exploration of more options of combinational targeted therapy in cancers may be the major challenge in the future.
Collapse
Affiliation(s)
- Jianjiang Fu
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China
| | - Xiaorui Su
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China
| | - Zhihua Li
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China
- Department of Fetal Medicine and Prenatal Diagnosis, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ling Deng
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiawei Liu
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China
| | - Xuancheng Feng
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China.
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China.
| | - Juan Peng
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China.
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China.
| |
Collapse
|
21
|
Land CA, Musich PR, Haydar D, Krenciute G, Xie Q. Chimeric antigen receptor T-cell therapy in glioblastoma: charging the T cells to fight. J Transl Med 2020; 18:428. [PMID: 33176788 PMCID: PMC7659102 DOI: 10.1186/s12967-020-02598-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant brain cancer that invades normal brain tissue and impedes surgical eradication, resulting in early local recurrence and high mortality. In addition, most therapeutic agents lack permeability across the blood brain barrier (BBB), further reducing the efficacy of chemotherapy. Thus, effective treatment against GBM requires tumor specific targets and efficient intracranial drug delivery. With the most recent advances in immunotherapy, genetically engineered T cells with chimeric antigen receptors (CARs) are becoming a promising approach for treating cancer. By transducing T lymphocytes with CAR constructs containing a tumor-associated antigen (TAA) recognition domain linked to the constant regions of a signaling T cell receptor, CAR T cells may recognize a predefined TAA with high specificity in a non-MHC restricted manner, and is independent of antigen processing. Active T cells can travel across the BBB, providing additional advantage for drug delivery and tumor targeting. Here we review the CAR design and technical innovations, the major targets that are in pre-clinical and clinical development with a focus on GBM, and multiple strategies developed to improve CAR T cell efficacy.
Collapse
Affiliation(s)
- Craig A. Land
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Phillip R. Musich
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Dalia Haydar
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Qian Xie
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| |
Collapse
|
22
|
Giotta Lucifero A, Luzzi S, Brambilla I, Schena L, Mosconi M, Foiadelli T, Savasta S. Potential roads for reaching the summit: an overview on target therapies for high-grade gliomas. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:61-78. [PMID: 32608376 PMCID: PMC7975828 DOI: 10.23750/abm.v91i7-s.9956] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022]
Abstract
Background: The tailored targeting of specific oncogenes represents a new frontier in the treatment of high-grade glioma in the pursuit of innovative and personalized approaches. The present study consists in a wide-ranging overview of the target therapies and related translational challenges in neuro-oncology. Methods: A review of the literature on PubMed/MEDLINE on recent advances concerning the target therapies for treatment of central nervous system malignancies was carried out. In the Medical Subject Headings, the terms “Target Therapy”, “Target drug” and “Tailored Therapy” were combined with the terms “High-grade gliomas”, “Malignant brain tumor” and “Glioblastoma”. Articles published in the last five years were further sorted, based on the best match and relevance. The ClinicalTrials.gov website was used as a source of the main trials, where the search terms were “Central Nervous System Tumor”, “Malignant Brain Tumor”, “Brain Cancer”, “Brain Neoplasms” and “High-grade gliomas”. Results: A total of 137 relevant articles and 79 trials were selected. Target therapies entailed inhibitors of tyrosine kinases, PI3K/AKT/mTOR pathway, farnesyl transferase enzymes, p53 and pRB proteins, isocitrate dehydrogenases, histone deacetylases, integrins and proteasome complexes. The clinical trials mostly involved combined approaches. They were phase I, II, I/II and III in 33%, 42%, 16%, and 9% of the cases, respectively. Conclusion: Tyrosine kinase and angiogenesis inhibitors, in combination with standard of care, have shown most evidence of the effectiveness in glioblastoma. Resistance remains an issue. A deeper understanding of the molecular pathways involved in gliomagenesis is the key aspect on which the translational research is focusing, in order to optimize the target therapies of newly diagnosed and recurrent brain gliomas. (www.actabiomedica.it)
Collapse
Affiliation(s)
- Alice Giotta Lucifero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
| | - Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Ilaria Brambilla
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Lucia Schena
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Mario Mosconi
- Orthopaedic and Traumatology Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
| | - Thomas Foiadelli
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Salvatore Savasta
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| |
Collapse
|
23
|
Hervieu A, Heuss SF, Zhang C, Barrow-McGee R, Joffre C, Ménard L, Clarke PA, Kermorgant S. A PI3K- and GTPase-independent Rac1-mTOR mechanism mediates MET-driven anchorage-independent cell growth but not migration. Sci Signal 2020; 13:eaba8627. [PMID: 32576681 PMCID: PMC7329383 DOI: 10.1126/scisignal.aba8627] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Receptor tyrosine kinases (RTKs) are often overexpressed or mutated in cancers and drive tumor growth and metastasis. In the current model of RTK signaling, including that of MET, downstream phosphatidylinositol 3-kinase (PI3K) mediates both cell proliferation and cell migration, whereas the small guanosine triphosphatase (GTPase) Rac1 mediates cell migration. However, in cultured NIH3T3 and glioblastoma cells, we found that class I PI3K mediated oncogenic MET-induced cell migration but not anchorage-independent growth. In contrast, Rac1 regulated both processes in distinct ways. Downstream of PI3K, Rac1 mediated cell migration through its GTPase activity, whereas independently of PI3K, Rac1 mediated anchorage-independent growth in a GTPase-independent manner through an adaptor function. Through its RKR motif, Rac1 formed a complex with the kinase mTOR to promote its translocation to the plasma membrane, where its activity promoted anchorage-independent growth of the cell cultures. Inhibiting mTOR with rapamycin suppressed the growth of subcutaneous MET-mutant cell grafts in mice, including that of MET inhibitor-resistant cells. These findings reveal a GTPase-independent role for Rac1 in mediating a PI3K-independent MET-to-mTOR pathway and suggest alternative or combined strategies that might overcome resistance to RTK inhibitors in patients with cancer.
Collapse
Affiliation(s)
- Alexia Hervieu
- Spatial Signalling Team, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
- Signal Transduction and Molecular Pharmacology Team, CRUK Cancer Therapeutics Unit, Division of Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Sara Farrah Heuss
- Spatial Signalling Team, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Chi Zhang
- Signal Transduction and Molecular Pharmacology Team, CRUK Cancer Therapeutics Unit, Division of Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Rachel Barrow-McGee
- Spatial Signalling Team, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Carine Joffre
- Spatial Signalling Team, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Ludovic Ménard
- Spatial Signalling Team, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Paul Andrew Clarke
- Signal Transduction and Molecular Pharmacology Team, CRUK Cancer Therapeutics Unit, Division of Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Stéphanie Kermorgant
- Spatial Signalling Team, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
24
|
Qin Y, Musket A, Kou J, Preiszner J, Tschida BR, Qin A, Land CA, Staal B, Kang L, Tanner K, Jiang Y, Schweitzer JB, Largaespada DA, Xie Q. Overexpression of HGF/MET axis along with p53 inhibition induces de novo glioma formation in mice. Neurooncol Adv 2020; 2:vdaa067. [PMID: 32642717 PMCID: PMC7332240 DOI: 10.1093/noajnl/vdaa067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Aberrant MET receptor tyrosine kinase (RTK) activation leads to invasive tumor growth in different types of cancer. Overexpression of MET and its ligand hepatocyte growth factor (HGF) occurs more frequently in glioblastoma (GBM) than in low-grade gliomas. Although we have shown previously that HGF-autocrine activation predicts sensitivity to MET tyrosine kinase inhibitors (TKIs) in GBM, whether it initiates tumorigenesis remains elusive. Methods Using a well-established Sleeping Beauty (SB) transposon strategy, we injected human HGF and MET cDNA together with a short hairpin siRNA against Trp53 (SB-hHgf.Met.ShP53) into the lateral ventricle of neonatal mice to induce spontaneous glioma initiation and characterized the tumors with H&E and immunohistochemistry analysis. Glioma sphere cells also were isolated for measuring the sensitivity to specific MET TKIs. Results Mixed injection of SB-hHgf.Met.ShP53 plasmids induced de novo glioma formation with invasive tumor growth accompanied by HGF and MET overexpression. While glioma stem cells (GSCs) are considered as the tumor-initiating cells in GBM, both SB-hHgf.Met.ShP53 tumor sections and glioma spheres harvested from these tumors expressed GSC markers nestin, GFAP, and Sox 2. Moreover, specific MET TKIs significantly inhibited tumor spheres' proliferation and MET/MAPK/AKT signaling. Conclusions Overexpression of the HGF/MET axis along with p53 attenuation may transform neural stem cells into GSCs, resulting in GBM formation in mice. These tumors are primarily driven by the MET RTK pathway activation and are sensitive to MET TKIs. The SB-hHgf.Met.ShP53 spontaneous mouse glioma model provides a useful tool for studying GBM tumor biology and MET-targeting therapeutics.
Collapse
Affiliation(s)
- Yuan Qin
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Anna Musket
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Jianqun Kou
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Johanna Preiszner
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Barbara R Tschida
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anna Qin
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Craig A Land
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Ben Staal
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Liang Kang
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Kirk Tanner
- National Brain Tumor Society, Newton, Massachusetts, USA
| | - Yong Jiang
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - John B Schweitzer
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - David A Largaespada
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Qian Xie
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| |
Collapse
|
25
|
Tabibkhooei A, Izadpanahi M, Arab A, Zare-Mirzaei A, Minaeian S, Rostami A, Mohsenian A. Profiling of novel circulating microRNAs as a non-invasive biomarker in diagnosis and follow-up of high and low-grade gliomas. Clin Neurol Neurosurg 2019; 190:105652. [PMID: 31896490 DOI: 10.1016/j.clineuro.2019.105652] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Glioblastoma (GBM) is the most common primary malignant neoplasm of the central nervous system (CNS). Despite the progress in therapeutic strategies such as surgical techniques, radiotherapy, chemotherapy, and targeted therapy, prognosis and therapeutically convenient monitoring tools in patients with GBM has not improved significantly up to now.Therefore, exosomal miRNAs as novel non-invasive biomarkers having high sensitivity and specificity are required to improve diagnosis and to develop new targeted therapy strategies for GBM patients. The aim of the present study was to investigate a novel miRNA signature as a predictive biomarker for diagnosis and measurement of response to therapeutic interventions in plasma of GBM patients versus traumatic brain injury and diffuse low-grade astrocytoma (LGA) patients. PATIENTS AND METHODS Plasma exosomal-microRNAs were isolated from GBM (n = 25), LGA (n = 25), and head trauma patients (n = 15) as non-glioma control from March 2017 to June 2018 in Department of Neurosurgery at Rasoul-e-Akram Hospital. Through a bioinformatics analysis, we used Miranda, TargetScan, mirBase, DIANA-microT-CDS, and KEGG database as well as microarray data analysis from GEO for microRNA candidates. Finally, miR-210, miR-185, miR-5194, and miR-449 were selected among those miRNAs because they were recorded to target the maximum number of genes in EGFR and c-MET signaling pathways. Then, exosomal microRNAs were extracted from plasma of patients and quantitated by locked nucleic acid real-time PCR in GBM, LGA, and trauma patients. RESULTS This result is the first report on the role of circulating miR-185, miR-449, and miR-5194 in GBM compared to LGA and trauma. The plasma expression of miR-210 as an oncogenic miR was upregulated in GBM and LGA groups (P < 0.0001). Otherwise, miR-185, miR-5194, and miR-449 were significantly downregulated (P ≤ 0.05) in GBM and LGA compared to trauma patients. There was no significant downregulation in the expression of miR-185 between GBM and LGA, while the expression of miR-5194 (P ≤ 0.05) and miR-449 (P ≤ 0.05) was significantly decreased in GBM patients compared with LGA. CONCLUSIONS These results indicate that the levels of miR-210, miR-449, and miR-5194 are a promising diagnostic and prognostic biomarker positively correlated with histopathological grade and invasiveness of GBM. These findings imply that circulating microRNA can be potentially used as novel biomarkers for glioma that might be beneficial in clinical management of glioma patients.
Collapse
Affiliation(s)
- Alireza Tabibkhooei
- Department of Neurosurgery, Iran University of Medical Sciences, Tehran, Iran.
| | - Maryam Izadpanahi
- Department of Neurosurgery, Iran University of Medical Sciences, Tehran, Iran.
| | - Abolfazl Arab
- Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Ali Zare-Mirzaei
- Department of Pathology, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Minaeian
- Antimicrobial Resistance Research Center, Institute of Immunology and Infection Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Rostami
- Department of Neurosurgery, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Mohsenian
- Department of Neurosurgery, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Wang K, Huang R, Wu C, Li G, Zhao Z, Hu H, Liu Y. Receptor tyrosine kinase expression in high-grade gliomas before and after chemoradiotherapy. Oncol Lett 2019; 18:6509-6515. [PMID: 31807171 PMCID: PMC6876328 DOI: 10.3892/ol.2019.11017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 03/28/2019] [Indexed: 01/23/2023] Open
Abstract
Glioma is the most common type of malignant brain tumor, and is characterized by invasive growth and chemoradiotherapy resistance. The following Cancer Genome Atlas mutation subtypes were identified in initial high-grade gliomas and recurrent gliomas treated by chemoradiotherapy: Isocitrate dehydrogenase 1/2 (IDH1/2) mutation, epidermal growth factor receptor variant III (EGFRvIII) mutation, tumor protein P53 mutation, PTEN mutation, O6-methylguanine-DNA methyltransferase promoter methylation and telomerase reverse transcriptase (TERT) mutation. The expression profile of 58 receptor tyrosine kinases (RTKs) were also examined. It was revealed that the proneural tumor subtype and IDH1/2 mutation are more frequent in recurrent tumors compared with initial tumors. Lower frequencies of the classical subtype, EGFRvIII mutation and TERT mutation were identified in recurrent tumors. A set of six RTK genes in which the level of expression was influenced by chemoradiotherapy was identified. Survival analysis revealed that the expression of several RTKs, including apoptosis-associated tyrosine kinase, fibroblast growth factor receptor 1 and insulin-like growth factor 1 receptor (IGF1R), was associated with patient survival. The stimulation of glioma cells by IGF1 in vitro was found to decreased the viability of the cells following treatment with temozolomide (TMZ). In addition, the expression level of IGF1R was increased in glioma cells treated with TMZ. These data suggest that altered RTK expression levels may influence the sensitivity of glioma to chemoradiotherapy.
Collapse
Affiliation(s)
- Kuanyu Wang
- Department of Gamma Knife Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing 100050, P.R. China
| | - Ruoyu Huang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing 100050, P.R. China
| | - Chenxing Wu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Guanzhang Li
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing 100050, P.R. China
| | - Zheng Zhao
- Department of Neuropathology, Beijing Neurosurgical Institute, Beijing 100050, P.R. China
| | - Huimin Hu
- Department of Neuropathology, Beijing Neurosurgical Institute, Beijing 100050, P.R. China
| | - Yanwei Liu
- Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
27
|
Zhang Y, Nguyen TTT, Shang E, Mela A, Humala N, Mahajan A, Zhao J, Shu C, Torrini C, Sanchez-Quintero MJ, Kleiner G, Bianchetti E, Westhoff MA, Quinzii CM, Karpel-Massler G, Bruce JN, Canoll P, Siegelin MD. MET Inhibition Elicits PGC1α-Dependent Metabolic Reprogramming in Glioblastoma. Cancer Res 2019; 80:30-43. [PMID: 31694905 DOI: 10.1158/0008-5472.can-19-1389] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/18/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022]
Abstract
The receptor kinase c-MET has emerged as a target for glioblastoma therapy. However, treatment resistance emerges inevitably. Here, we performed global metabolite screening with metabolite set enrichment coupled with transcriptome and gene set enrichment analysis and proteomic screening, and identified substantial reprogramming of tumor metabolism involving oxidative phosphorylation and fatty acid oxidation (FAO) with substantial accumulation of acyl-carnitines accompanied by an increase of PGC1α in response to genetic (shRNA and CRISPR/Cas9) and pharmacologic (crizotinib) inhibition of c-MET. Extracellular flux and carbon tracing analyses (U-13C-glucose, U-13C-glutamine, and U-13C-palmitic acid) demonstrated enhanced oxidative metabolism, which was driven by FAO and supported by increased anaplerosis of glucose carbons. These findings were observed in concert with increased number and fusion of mitochondria and production of reactive oxygen species. Genetic interference with PGC1α rescued this oxidative phenotype driven by c-MET inhibition. Silencing and chromatin immunoprecipitation experiments demonstrated that cAMP response elements binding protein regulates the expression of PGC1α in the context of c-MET inhibition. Interference with both oxidative phosphorylation (metformin, oligomycin) and β-oxidation of fatty acids (etomoxir) enhanced the antitumor efficacy of c-MET inhibition. Synergistic cell death was observed with c-MET inhibition and gamitrinib treatment. In patient-derived xenograft models, combination treatments of crizotinib and etomoxir, and crizotinib and gamitrinib were significantly more efficacious than single treatments and did not induce toxicity. Collectively, we have unraveled the mechanistic underpinnings of c-MET inhibition and identified novel combination therapies that may enhance its therapeutic efficacy. SIGNIFICANCE: c-MET inhibition causes profound metabolic reprogramming that can be targeted by drug combination therapies.
Collapse
Affiliation(s)
- Yiru Zhang
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Trang T T Nguyen
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Enyuan Shang
- Department of Biological Sciences, Bronx Community College, City University of New York, Bronx, New York
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Nelson Humala
- Department of Neurological Surgery, Columbia University Medical Center, New York, New York
| | - Aayushi Mahajan
- Department of Neurological Surgery, Columbia University Medical Center, New York, New York
| | - Junfei Zhao
- Department of Biomedical Informatics, Columbia University, New York, New York
| | - Chang Shu
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Consuelo Torrini
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | | | - Giulio Kleiner
- Department of Neurology, Columbia University Medical Center, New York, New York
| | - Elena Bianchetti
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Catarina M Quinzii
- Department of Neurology, Columbia University Medical Center, New York, New York
| | | | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Medical Center, New York, New York
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York.
| |
Collapse
|
28
|
Cheng F, Guo D. MET in glioma: signaling pathways and targeted therapies. J Exp Clin Cancer Res 2019; 38:270. [PMID: 31221203 PMCID: PMC6585013 DOI: 10.1186/s13046-019-1269-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Gliomas represent the most common type of malignant brain tumor, among which, glioblastoma remains a clinical challenge with limited treatment options and dismal prognosis. It has been shown that the dysregulated receptor tyrosine kinase (RTK, including EGFR, MET, PDGFRα, ect.) signaling pathways have pivotal roles in the progression of gliomas, especially glioblastoma. Increasing evidence suggests that expression levels of the RTK MET and its specific stimulatory factors are significantly increased in glioblastomas compared to those in normal brain tissues, whereas some negative regulators are found to be downregulated. Mutations in MET, as well as the dysregulation of other regulators of cross-talk with MET signaling pathways, have also been identified. MET and its ligand hepatocyte growth factor (HGF) play a critical role in the proliferation, survival, migration, invasion, angiogenesis, stem cell characteristics, and therapeutic resistance and recurrence of glioblastomas. Therefore, combined targeted therapy for this pathway and associated molecules could be a novel and attractive strategy for the treatment of human glioblastoma. In this review, we highlight progress made in the understanding of MET signaling in glioma and advances in therapies targeting HGF/MET molecules for glioma patients in recent years, in addition to studies on the expression and mutation status of MET.
Collapse
Affiliation(s)
- Fangling Cheng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, 430030 China
| | - Dongsheng Guo
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, 430030 China
| |
Collapse
|
29
|
Cheng F, Zhang P, Xiao Q, Li Y, Dong M, Wang H, Kuang D, He Y, Duan Q, Mao F, Wang B, Guo D. The Prognostic and Therapeutic Potential of LRIG3 and Soluble LRIG3 in Glioblastoma. Front Oncol 2019; 9:447. [PMID: 31245283 PMCID: PMC6563081 DOI: 10.3389/fonc.2019.00447] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/13/2019] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma is a highly lethal type of primary brain tumor that exhibits unrestricted growth and aggressive invasion capabilities, leading to a dismal prognosis despite a multitude of therapies. Multiple alterations in the expression level of genes and/or proteins have been identified in glioblastomas, including the activation of oncogenes and/or silencing of tumor-suppressor genes. Nevertheless, there are still no effective targeted therapies associated with these changes. In this study, we investigated the expression of human leucine-rich repeats and immunoglobulin-like domains protein 3 (LRIG3) in human glioma specimens through immunohistochemical analysis. The results showed that LRIG3 was weakly expressed in high-grade gliomas (WHO [World Health Organization] grades III and IV) compared with that in low-grade gliomas (WHO grade II). Survival analysis of these patients with glioma indicated that LRIG3 is an important prognostic marker for better survival. Moreover, we confirmed the existence of soluble ectodomain of LRIG3 (sLRIG3) in the cell culture supernatant, serum, and in tumor cystic fluid of patients with glioma. Molecular mechanistic investigation demonstrated that both LRIG3 and sLRIG3 inhibit the growth and invasion capabilities of GL15, U87, and PriGBM cells and tumor xenografts in nude mice through regulating the MET/phosphatidylinositol 3-kinase/Akt signaling pathway. Enzyme-linked immunosorbent assay confirmed the positive correlation between serum sLRIG3 protein levels and overall survival time in patients with high-grade gliomas. Taken together, our data for the first time demonstrate the existence of sLRIG3 and that both LRIG3 and sLRIG3 are potent tumor suppressors, which could be used as prognostic markers for better overall survival and therapeutic agents for glioblastoma.
Collapse
Affiliation(s)
- Fangling Cheng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Chinese-German Lab of Molecular Neuro-oncology of Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Po Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Chinese-German Lab of Molecular Neuro-oncology of Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qungen Xiao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Youwei Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Chinese-German Lab of Molecular Neuro-oncology of Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Minhai Dong
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Chinese-German Lab of Molecular Neuro-oncology of Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Heping Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Kuang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue He
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuhong Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Mao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baofeng Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongsheng Guo
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
SPINT2 is hypermethylated in both IDH1 mutated and wild-type glioblastomas, and exerts tumor suppression via reduction of c-Met activation. J Neurooncol 2019; 142:423-434. [PMID: 30838489 DOI: 10.1007/s11060-019-03126-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/09/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Both IDH1-mutated and wild-type gliomas abundantly display aberrant CpG island hypermethylation. However, the potential role of hypermethylation in promoting gliomas, especially the most aggressive form, glioblastoma (GBM), remains poorly understood. METHODS We analyzed RRBS-generated methylation profiles for 11 IDH1WT gliomas (including 7 GBMs), 24 IDH1MUT gliomas (including 6 GBMs), and 5 normal brain samples and employed TCGA GBM methylation profiles as a validation set. Upon classification of differentially methylated CpG islands by IDH1 status, we used integrated analysis of methylation and gene expression to identify SPINT2 as a top cancer related gene. To explore functional consequences of SPINT2 methylation in GBM, we validated SPINT2 methylation status using targeted bisulfite sequencing in a large cohort of GBM samples. We assessed DNA methylation-mediated SPINT2 gene regulation using 5-aza-2'-deoxycytidine treatment, DNMT1 knockdown and luciferase reporter assays. We conducted functional analyses of SPINT2 in GBM cell lines in vitro and in vivo. RESULTS We identified SPINT2 as a candidate tumor-suppressor gene within a group of CpG islands (designated GT-CMG) that are hypermethylated in both IDH1MUT and IDH1WT gliomas but not in normal brain. We established that SPINT2 downregulation results from promoter hypermethylation, and that restoration of SPINT2 expression reduces c-Met activation and tumorigenic properties of GBM cells. CONCLUSIONS We defined a previously under-recognized group of coordinately methylated CpG islands common to both IDH1WT and IDH1MUT gliomas (GT-CMG). Within GT-CMG, we identified SPINT2 as a top cancer-related candidate and demonstrated that SPINT2 suppressed GBM via down-regulation of c-Met activation.
Collapse
|
31
|
Baltschukat S, Engstler BS, Huang A, Hao HX, Tam A, Wang HQ, Liang J, DiMare MT, Bhang HEC, Wang Y, Furet P, Sellers WR, Hofmann F, Schoepfer J, Tiedt R. Capmatinib (INC280) Is Active Against Models of Non–Small Cell Lung Cancer and Other Cancer Types with Defined Mechanisms of MET Activation. Clin Cancer Res 2019; 25:3164-3175. [DOI: 10.1158/1078-0432.ccr-18-2814] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/12/2018] [Accepted: 01/18/2019] [Indexed: 11/16/2022]
|
32
|
Hughes VS, Siemann DW. Have Clinical Trials Properly Assessed c-Met Inhibitors? Trends Cancer 2018; 4:94-97. [PMID: 29458966 DOI: 10.1016/j.trecan.2017.11.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/27/2017] [Accepted: 11/30/2017] [Indexed: 01/03/2023]
Abstract
The c-Met/HGF pathway is implicated in cancer progression and dissemination. Many inhibitors have been developed to target this pathway. Unfortunately, most trials have failed to demonstrate efficacy. However, clinical trials have not adequately tested the concept of c-Met pathway inhibition due to the lack of appropriate patient selection criteria.
Collapse
Affiliation(s)
- Veronica S Hughes
- Department of Radiation Oncology, University of Florida, 2033 Mowry Road, Cancer Genetic Research Complex, Room 485E, Gainesville, FL 32610, USA.
| | - Dietmar W Siemann
- Department of Radiation Oncology, University of Florida, 2033 Mowry Road, Cancer Genetic Research Complex, Room 485E, Gainesville, FL 32610, USA
| |
Collapse
|
33
|
HGF/c-MET Signaling in Melanocytes and Melanoma. Int J Mol Sci 2018; 19:ijms19123844. [PMID: 30513872 PMCID: PMC6321285 DOI: 10.3390/ijms19123844] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 12/17/2022] Open
Abstract
Hepatocyte growth factor (HGF)/ mesenchymal-epithelial transition factor (c-MET) signaling is involved in complex cellular programs that are important for embryonic development and tissue regeneration, but its activity is also utilized by cancer cells during tumor progression. HGF and c-MET usually mediate heterotypic cell–cell interactions, such as epithelial–mesenchymal, including tumor–stroma interactions. In the skin, dermal fibroblasts are the main source of HGF. The presence of c-MET on keratinocytes is crucial for wound healing in the skin. HGF is not released by normal melanocytes, but as melanocytes express c-MET, they are receptive to HGF, which protects them from apoptosis and stimulates their proliferation and motility. Dissimilar to melanocytes, melanoma cells not only express c-MET, but also release HGF, thus activating c-MET in an autocrine manner. Stimulation of the HGF/c-MET pathways contributes to several processes that are crucial for melanoma development, such as proliferation, survival, motility, and invasiveness, including distant metastatic niche formation. HGF might be a factor in the innate and acquired resistance of melanoma to oncoprotein-targeted drugs. It is not entirely clear whether elevated serum HGF level is associated with low progression-free survival and overall survival after treatment with targeted therapies. This review focuses on the role of HGF/c-MET signaling in melanoma with some introductory information on its function in skin and melanocytes.
Collapse
|
34
|
Nickel AC, Wan XY, Saban DV, Weng YL, Zhang S, Keyvani K, Sure U, Zhu Y. Loss of programmed cell death 10 activates tumor cells and leads to temozolomide-resistance in glioblastoma. J Neurooncol 2018; 141:31-41. [PMID: 30392087 DOI: 10.1007/s11060-018-03017-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/25/2018] [Indexed: 11/24/2022]
Abstract
PURPOSE Glioblastoma (GBM) is one of the most aggressive and incurable primary brain tumors. Identification of novel therapeutic targets is an urgent priority. Programmed cell death 10 (PDCD10), a ubiquitously expressed apoptotic protein, has shown a dual function in different types of cancers and in chemo-resistance. Recently, we reported that PDCD10 was downregulated in human GBM. The aim of this study was to explore the function of PDCD10 in GBM cells. METHODS PDCD10 was knocked down in three GBM cell lines (U87, T98g and LN229) by lentiviral-mediated shRNA transduction. U87 and T98g transduced cells were used for phenotype study and LN229 and T98g cells were used for apoptosis study. The role of PDCD10 in apoptosis and chemo-resistance was investigated after treatment with staurosporine and temozolomide. A GBM xenograft mouse model was used to confirm the function of PDCD10 in vivo. A protein array was performed in PDCD10-knockdown and control GBM cells. RESULTS Knockdown of PDCD10 in GBM cells promoted cell proliferation, adhesion, migration, invasion, and inhibited apoptosis and caspase-3 activation. PDCD10-knockdown accelerated tumor growth and increased tumor mass by 2.1-fold and led to a chemo-resistance of mice treated with temozolomide. Immunostaining revealed extensive Ki67-positive cells and less activation of caspase-3 in PDCD10-knockdown tumors. The protein array demonstrated an increased release of multiple growth factors from PDCD10-knockdown GBM cells. CONCLUSIONS Loss of programmed cell death 10 activates tumor cells and leads to temozolomide-resistance in GBM, suggesting PDCD10 as a potential target for GBM therapy.
Collapse
Affiliation(s)
- Ann-Christin Nickel
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Xue-Yan Wan
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.,Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dino-Vitali Saban
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Yin-Lun Weng
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.,Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shu Zhang
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Ulrich Sure
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Yuan Zhu
- Department of Neurosurgery, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|
35
|
Noriega-Guerra H, Freitas VM. Extracellular Matrix Influencing HGF/c-MET Signaling Pathway: Impact on Cancer Progression. Int J Mol Sci 2018; 19:ijms19113300. [PMID: 30352967 PMCID: PMC6274944 DOI: 10.3390/ijms19113300] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/18/2018] [Accepted: 10/20/2018] [Indexed: 12/22/2022] Open
Abstract
The extracellular matrix (ECM) is a crucial component of the tumor microenvironment involved in numerous cellular processes that contribute to cancer progression. It is acknowledged that tumor–stromal cell communication is driven by a complex and dynamic network of cytokines, growth factors and proteases. Thus, the ECM works as a reservoir for bioactive molecules that modulate tumor cell behavior. The hepatocyte growth factor (HGF) produced by tumor and stromal cells acts as a multifunctional cytokine and activates the c-MET receptor, which is expressed in different tumor cell types. The HGF/c-MET signaling pathway is associated with several cellular processes, such as proliferation, survival, motility, angiogenesis, invasion and metastasis. Moreover, c-MET activation can be promoted by several ECM components, including proteoglycans and glycoproteins that act as bridging molecules and/or signal co-receptors. In contrast, c-MET activation can be inhibited by proteoglycans, matricellular proteins and/or proteases that bind and sequester HGF away from the cell surface. Therefore, understanding the effects of ECM components on HGF and c-MET may provide opportunities for novel therapeutic strategies. Here, we give a short overview of how certain ECM components regulate the distribution and activation of HGF and c-MET.
Collapse
Affiliation(s)
- Heydi Noriega-Guerra
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes 1524, Prédio I, sala 428, 05508-000, São Paulo, SP, Brazil.
| | - Vanessa Morais Freitas
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes 1524, Prédio I, sala 428, 05508-000, São Paulo, SP, Brazil.
| |
Collapse
|
36
|
Kou J, Musich PR, Staal B, Kang L, Qin Y, Yao ZQ, Zhang B, Wu W, Tam A, Huang A, Hao HX, Vande Woude GF, Xie Q. Differential responses of MET activations to MET kinase inhibitor and neutralizing antibody. J Transl Med 2018; 16:253. [PMID: 30208970 PMCID: PMC6134500 DOI: 10.1186/s12967-018-1628-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Aberrant MET tyrosine kinase signaling is known to cause cancer initiation and progression. While MET inhibitors are in clinical trials against several cancer types, the clinical efficacies are controversial and the molecular mechanisms toward sensitivity remain elusive. METHODS With the goal to investigate the molecular basis of MET amplification (METamp) and hepatocyte growth factor (HGF) autocrine-driven tumors in response to MET tyrosine kinase inhibitors (TKI) and neutralizing antibodies, we compared cancer cells harboring METamp (MKN45 and MHCCH97H) or HGF-autocrine (JHH5 and U87) for their sensitivity and downstream biological responses to a MET-TKI (INC280) and an anti-MET monoclonal antibody (MetMab) in vitro, and for tumor inhibition in vivo. RESULTS We find that cancer cells driven by METamp are more sensitive to INC280 than are those driven by HGF-autocrine activation. In METamp cells, INC280 induced a DNA damage response with activation of repair through the p53BP1/ATM signaling pathway. Although MetMab failed to inhibit METamp cell proliferation and tumor growth, both INC280 and MetMab reduced HGF-autocrine tumor growth. In addition, we also show that HGF stimulation promoted human HUVEC cell tube formation via the Src pathway, which was inhibited by either INC280 or MetMab. These observations suggest that in HGF-autocrine tumors, the endothelial cells are the secondary targets MET inhibitors. CONCLUSIONS Our results demonstrate that METamp and HGF-autocrine activation favor different molecular mechanisms. While combining MET TKIs and ATM inhibitors may enhance the efficacy for treating tumors harboring METamp, a combined inhibition of MET and angiogenesis pathways may improve the therapeutic efficacy against HGF-autocrine tumors.
Collapse
Affiliation(s)
- Jianqun Kou
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Phillip R Musich
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Ben Staal
- Center of Cell and Cancer Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Liang Kang
- Center of Cell and Cancer Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Yuan Qin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Zhi Q Yao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.,Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Boheng Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Weizhong Wu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Angela Tam
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - Alan Huang
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - Huai-Xiang Hao
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - George F Vande Woude
- Center of Cell and Cancer Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Qian Xie
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA. .,Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
| |
Collapse
|
37
|
Fukushima T, Kawaguchi M, Yamamoto K, Yamashita F, Izumi A, Kaieda T, Takezaki Y, Itoh H, Takeshima H, Kataoka H. Aberrant methylation and silencing of the SPINT2 gene in high-grade gliomas. Cancer Sci 2018; 109:2970-2979. [PMID: 29987920 PMCID: PMC6125435 DOI: 10.1111/cas.13732] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/03/2018] [Accepted: 07/08/2018] [Indexed: 12/25/2022] Open
Abstract
Hepatocyte growth factor activator inhibitor type 2 (HAI‐2), encoded by the SPINT2 gene, is a membrane‐anchored protein that inhibits proteases involved in the activation of hepatocyte growth factor (HGF), a ligand of MET receptor. Epigenetic silencing of the SPINT2 gene has been reported in a human glioblastoma cell line (U87) and glioblastoma‐derived cancer stem cells. However, the incidence of SPINT2 methylation in tumor tissues obtained from glioma patients is unknown. In this study, we analyzed the methylation status of the SPINT2 gene of eight human glioblastoma cell lines and surgically resected glioma tissues of different grades (II, III, and IV) by bisulfite sequence analysis and methylation‐specific PCR. Most glioblastoma lines (7/8) showed methylation of the SPINT2 gene with a significantly reduced level of SPINT2mRNA compared to cultured astrocytes and normal brain tissues. However, all glioblastoma lines expressed mRNA for HGF activator (HGFAC), a target protease of HAI‐2/SPINT2. Forced expression of SPINT2 reduced MET phosphorylation of U87 glioblastoma cells both in vitro and in intracranial xenografts in nude mice. Methylation‐specific PCR analysis of the resected glioma tissues indicated notable methylation of the SPINT2 gene in 33.3% (2/6), 71.4% (10/14), and 74.3% (26/35) of grade II, III, and IV gliomas, respectively. Analysis of RNA sequencing data in a public database indicated an increased HGFAC/SPINT2 expression ratio in high‐grade compared to low‐grade gliomas (P = .01). In summary, aberrant methylation of the SPINT2 gene is frequently observed in high‐grade gliomas and might confer MET signaling in the glioma cells.
Collapse
Affiliation(s)
- Tsuyoshi Fukushima
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Makiko Kawaguchi
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Koji Yamamoto
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Fumiki Yamashita
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Aya Izumi
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Takashi Kaieda
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yuka Takezaki
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hiroshi Itoh
- Department of Molecular Pathology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Hideo Takeshima
- Section of Neurosurgery, Department of Clinical Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hiroaki Kataoka
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
38
|
Hypoxia leads to decreased autophosphorylation of the MET receptor but promotes its resistance to tyrosine kinase inhibitors. Oncotarget 2018; 9:27039-27058. [PMID: 29930749 PMCID: PMC6007473 DOI: 10.18632/oncotarget.25472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 05/08/2018] [Indexed: 12/16/2022] Open
Abstract
The receptor tyrosine kinase MET and its ligand, the Hepatocyte Growth Factor/Scattor Factor (HGF/SF), are essential to the migration, morphogenesis, and survival of epithelial cells. In addition, dysregulation of MET signaling has been shown to promote tumor progression and invasion in many cancers. Therefore, HGF/SF and MET are major targets for chemotherapies. Improvement of targeted therapies requires a perfect understanding of tumor microenvironment that strongly modifies half-life, bio-accessibility and thus, efficacy of treatments. In particular, hypoxia is a crucial microenvironmental phenomenon promoting invasion and resistance to treatments. Under hypoxia, MET auto-phosphorylation resulting from ligand stimulation or from receptor overexpression is drastically decreased within minutes of oxygen deprivation but is quickly reversible upon return to normoxia. Besides a decreased phosphorylation of its proximal adaptor GAB1 under hypoxia, activation of the downstream kinases Erk and Akt is maintained, while still being dependent on MET receptor. Consistently, several cellular responses induced by HGF/SF, including motility, morphogenesis, and survival are effectively induced under hypoxia. Interestingly, using a semi-synthetic ligand, we show that HGF/SF binding to MET is strongly impaired during hypoxia but can be quickly restored upon reoxygenation. Finally, we show that two MET-targeting tyrosine kinase inhibitors (TKIs) are less efficient on MET signalling under hypoxia. Like MET loss of phosphorylation, this hypoxia-induced resistance to TKIs is reversible under normoxia. Thus, although hypoxia does not affect downstream signaling or cellular responses induced by MET, it causes immediate resistance to TKIs. These results may prove useful when designing and evaluation of MET-targeted therapies against cancer.
Collapse
|
39
|
Raghav K, Bailey AM, Loree JM, Kopetz S, Holla V, Yap TA, Wang F, Chen K, Salgia R, Hong D. Untying the gordion knot of targeting MET in cancer. Cancer Treat Rev 2018; 66:95-103. [PMID: 29730462 DOI: 10.1016/j.ctrv.2018.04.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 01/30/2023]
Abstract
Despite compelling evidence backing the crucial role of a dysregulated MET axis in cancer and a myriad of agents targeting this pathway in active clinical development, the therapeutic value of MET inhibition in cancer oncology remains to be established. Although a series of disappointing clinical trials, at first, lessened fervor for targeting this pathway, investigations continue unabated with a number of novel active compounds entering clinical trials. Suboptimal designs which lacked biomarker selection have been the main reason for these early failures and this has stimulated a more biomarker enriched approach lately. Fresh insights into the mechanics of diverse MET aberrations (amplifications and mutations) have allowed trial enrichment for appropriate patients in appropriate disease settings. Development of MET inhibition as a therapeutic strategy in cancer has been a lesson in itself reflecting the challenging opportunities enclosed in the genetic landscape of cancer. Here, we will review the status of MET targeted therapy in development as it stands today, discuss emerging paradigms in MET inhibition and theorize on concepts for future development. We venture to propose that in spite of early disappointments, the future of this therapeutic strategy is promising with use of appropriate predictive biomarker in the right clinical context.
Collapse
Affiliation(s)
- Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ann Marie Bailey
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jonathan M Loree
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vijaykumar Holla
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Timothy Anthony Yap
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Fang Wang
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ken Chen
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - David Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
40
|
The multiple paths towards MET receptor addiction in cancer. Oncogene 2018; 37:3200-3215. [PMID: 29551767 DOI: 10.1038/s41388-018-0185-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/30/2018] [Accepted: 01/30/2018] [Indexed: 12/14/2022]
Abstract
Targeted therapies against receptor tyrosine kinases (RTKs) are currently used with success on a small proportion of patients displaying clear oncogene activation. Lung cancers with a mutated EGFR provide a good illustration. The efficacy of targeted treatments relies on oncogene addiction, a situation in which the growth or survival of the cancer cells depends on a single deregulated oncogene. MET, a member of the RTK family, is a promising target because it displays many deregulations in a broad panel of cancers. Although clinical trials having evaluated MET inhibitors in large populations have yielded disappointing results, many recent case reports suggest that MET inhibition may be effective in a subset of patients with unambiguous MET activation and thus, most probably, oncogene addiction. Interestingly, preclinical studies have revealed a particularity of MET addiction: it can arise through several mechanisms, and the mechanism involved can differ according to the cancer type. The present review describes the different mechanisms of MET addiction and their consequences for diagnosis and therapeutic strategies. Although in each cancer type MET addiction affects a restricted number of patients, pooling of these patients across all cancer types yields a targetable population liable to benefit from addiction-targeting therapies.
Collapse
|
41
|
Fiedler U, Ekawardhani S, Cornelius A, Gilboy P, Bakker TR, Dolado I, Stumpp MT, Dawson KM. MP0250, a VEGF and HGF neutralizing DARPin ® molecule shows high anti-tumor efficacy in mouse xenograft and patient-derived tumor models. Oncotarget 2017; 8:98371-98383. [PMID: 29228696 PMCID: PMC5716736 DOI: 10.18632/oncotarget.21738] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/21/2017] [Indexed: 01/13/2023] Open
Abstract
Background The VEGF/VEGFR and the HGF/cMET pathways are key mediators of the interplay of tumor cells and their microenvironment. However, inhibition of VEGF has been shown to produce only limited clinical benefit and inhibition of the activation of cMET by HGF has not translated into clinical benefit in pivotal trials. MP0250, a DARPin® molecule that specifically inhibits both VEGF and HGF has been developed to explore the clinical potential of dual inhibition of these pathways. Results MP0250 binding to VEGF and HGF inhibited downstream signalling through VEGFR2 and cMET resulting in inhibition of proliferation of VEGF- and HGF-dependent cells. Antitumor activity was demonstrated in VEGF- and HGF-dependent xenograft and syngeneic models with activity superior to that of individual VEGF- and HGF-blocking DARPin® molecules. Combination therapy studies showed potentiation of the antitumor activity of chemotherapy and immunotherapy agents, including an anti-PD1 antibody. Materials and Methods Potency of MP0250 was assessed in cellular models and in a variety of xenograft models as monotherapy or in combination with standard-of-care drugs. Conclusions Dual inhibition of VEGF and HGF by MP0250 produced powerful single agent and combination antitumor activity. This, together with increasing understanding of the role of the HGF/cMET pathway in resistance to VEGF (and other agents), supports testing of MP0250 in the clinic.
Collapse
Affiliation(s)
| | | | | | - Pat Gilboy
- Molecular Partners AG, Schlieren, Switzerland.,Horizon Pharma, Dublin, Ireland
| | - Talitha R Bakker
- Molecular Partners AG, Schlieren, Switzerland.,Amgen (Europe) GmbH, Zug, Switzerland
| | - Ignacio Dolado
- Molecular Partners AG, Schlieren, Switzerland.,Roche, Basel, Switzerland
| | | | | |
Collapse
|
42
|
Wierzbicki M, Jaworski S, Kutwin M, Grodzik M, Strojny B, Kurantowicz N, Zdunek K, Chodun R, Chwalibog A, Sawosz E. Diamond, graphite, and graphene oxide nanoparticles decrease migration and invasiveness in glioblastoma cell lines by impairing extracellular adhesion. Int J Nanomedicine 2017; 12:7241-7254. [PMID: 29042773 PMCID: PMC5634373 DOI: 10.2147/ijn.s146193] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The highly invasive nature of glioblastoma is one of the most significant problems regarding the treatment of this tumor. Diamond nanoparticles (ND), graphite nanoparticles (NG), and graphene oxide nanoplatelets (nGO) have been explored for their biomedical applications, especially for drug delivery. The objective of this research was to assess changes in the adhesion, migration, and invasiveness of two glioblastoma cell lines, U87 and U118, after ND, NG, and nGO treatment. All treatments affected the cell surface structure, adhesion-dependent EGFR/AKT/mTOR, and β-catenin signaling pathways, decreasing the migration and invasiveness of both glioblastoma cell lines. The examined nanoparticles did not show strong toxicity but effectively deregulated cell migration. ND was effectively taken up by cells, whereas nGO and NG strongly interacted with the cell surface. These results indicate that nanoparticles could be used in biomedical applications as a low toxicity active compound for glioblastoma treatment.
Collapse
Affiliation(s)
| | | | - Marta Kutwin
- Division of Nanobiotechnology, Warsaw University of Life Science
| | - Marta Grodzik
- Division of Nanobiotechnology, Warsaw University of Life Science
| | - Barbara Strojny
- Division of Nanobiotechnology, Warsaw University of Life Science
| | | | - Krzysztof Zdunek
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Rafał Chodun
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - André Chwalibog
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ewa Sawosz
- Division of Nanobiotechnology, Warsaw University of Life Science
| |
Collapse
|
43
|
Tsuji T, Sakamori Y, Ozasa H, Yagi Y, Ajimizu H, Yasuda Y, Funazo T, Nomizo T, Yoshida H, Nagai H, Maeno K, Oguri T, Hirai T, Kim YH. Clinical impact of high serum hepatocyte growth factor in advanced non-small cell lung cancer. Oncotarget 2017; 8:71805-71816. [PMID: 29069748 PMCID: PMC5641091 DOI: 10.18632/oncotarget.17895] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/29/2017] [Indexed: 12/17/2022] Open
Abstract
Activation of c-MET through hepatocyte growth factor (HGF) increases tumorigenesis, induces resistance, and is associated with poor prognosis in various solid tumors. However, the clinical value of serum HGF (sHGF) in patients with advanced non-small cell lung cancer (NSCLC), especially those receiving cytotoxic chemotherapy, remains unknown. Here, we show that sHGF may be useful to predict tumor response and progression-free survival (PFS) in patients with advanced NSCLC. A total of 81 patients with NSCLC were investigated. sHGF levels were evaluated using ELISA at 4 time-points: at pre-treatment, at response-evaluation (1-2 months after treatment initiation), at the best tumor response, and at disease progression. As a control biomarker, CEA was also evaluated in lung adenocarcinoma. Positive-sHGF at response-evaluation predicted poor PFS compared with Negative-sHGF in both first-line (median, 153.5 vs. 288.0; P < 0.05) and second-line treatment (87.0 vs. 219.5; P = 0.01). In 55 patients that received cytotoxic chemotherapy, multiple Cox proportional hazards models showed significant independent associations between poor PFS and Positive-sHGF at response-evaluation (hazard ratio, 4.24; 95% CI, 2.05 to 9.46; P < 0.01). Lung adenocarcinoma subgroup analysis showed that in patients receiving second cytotoxic chemotherapy, there were no significant differences in PFS between patients with low-CEA compared with those with high-CEA, but Positive-sHGF at pre-treatment or at response-evaluation predicted poor PFS (35.0 vs. 132.0; P < 0.01, 50.0 vs. 215.0; P < 0.01, respectively). These findings give a rationale for future research investigating the merit of sHGF as a potential clinical biomarker to evaluate HGF/c-MET activity, which would be useful to indicate administration of c-MET inhibitors.
Collapse
Affiliation(s)
- Takahiro Tsuji
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuichi Sakamori
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroaki Ozasa
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshitaka Yagi
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hitomi Ajimizu
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuto Yasuda
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoko Funazo
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Nomizo
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hironori Yoshida
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroki Nagai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Maeno
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tetsuya Oguri
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Young Hak Kim
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
44
|
Clémenson C, Chargari C, Liu W, Mondini M, Ferté C, Burbridge MF, Cattan V, Jacquet-Bescond A, Deutsch E. The MET/AXL/FGFR Inhibitor S49076 Impairs Aurora B Activity and Improves the Antitumor Efficacy of Radiotherapy. Mol Cancer Ther 2017; 16:2107-2119. [PMID: 28619752 DOI: 10.1158/1535-7163.mct-17-0112] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/21/2017] [Accepted: 06/05/2017] [Indexed: 11/16/2022]
Abstract
Several therapeutic agents targeting HGF/MET signaling are under clinical development as single agents or in combination, notably with anti-EGFR therapies in non-small cell lung cancer (NSCLC). However, despite increasing data supporting a link between MET, irradiation, and cancer progression, no data regarding the combination of MET-targeting agents and radiotherapy are available from the clinic. S49076 is an oral ATP-competitive inhibitor of MET, AXL, and FGFR1-3 receptors that is currently in phase I/II clinical trials in combination with gefitinib in NSCLC patients whose tumors show resistance to EGFR inhibitors. Here, we studied the impact of S49076 on MET signaling, cell proliferation, and clonogenic survival in MET-dependent (GTL16 and U87-MG) and MET-independent (H441, H460, and A549) cells. Our data show that S49076 exerts its cytotoxic activity at low doses on MET-dependent cells through MET inhibition, whereas it inhibits growth of MET-independent cells at higher but clinically relevant doses by targeting Aurora B. Furthermore, we found that S49076 improves the antitumor efficacy of radiotherapy in both MET-dependent and MET-independent cell lines in vitro and in subcutaneous and orthotopic tumor models in vivo In conclusion, our study demonstrates that S49076 has dual antitumor activity and can be used in combination with radiotherapy for the treatment of both MET-dependent and MET-independent tumors. These results support the evaluation of combined treatment of S49076 with radiation in clinical trials without patient selection based on the tumor MET dependency status. Mol Cancer Ther; 16(10); 2107-19. ©2017 AACR.
Collapse
Affiliation(s)
- Céline Clémenson
- Gustave Roussy, Université Paris-Saclay, UMR Radiothérapie Moléculaire, Villejuif, France.,INSERM, U1030, SIRIC Socrates, DHU TORINO, Villejuif, France
| | - Cyrus Chargari
- Gustave Roussy, Université Paris-Saclay, UMR Radiothérapie Moléculaire, Villejuif, France.,INSERM, U1030, SIRIC Socrates, DHU TORINO, Villejuif, France.,Gustave Roussy, Université Paris-Saclay, Département de Radiothérapie, Villejuif, France.,Institut de Recherche Biomédicale des Armées, Brétigny-Sur-Orge, France
| | - Winchygn Liu
- Gustave Roussy, Université Paris-Saclay, UMR Radiothérapie Moléculaire, Villejuif, France.,INSERM, U1030, SIRIC Socrates, DHU TORINO, Villejuif, France
| | - Michele Mondini
- Gustave Roussy, Université Paris-Saclay, UMR Radiothérapie Moléculaire, Villejuif, France.,INSERM, U1030, SIRIC Socrates, DHU TORINO, Villejuif, France
| | - Charles Ferté
- Gustave Roussy, Université Paris-Saclay, UMR Radiothérapie Moléculaire, Villejuif, France.,INSERM, U1030, SIRIC Socrates, DHU TORINO, Villejuif, France.,INSERM, U981, Villejuif, France
| | - Mike F Burbridge
- Oncology Unit, Institut de Recherches Internationales Servier, Suresnes, France
| | - Valérie Cattan
- Oncology Unit, Institut de Recherches Internationales Servier, Suresnes, France
| | | | - Eric Deutsch
- Gustave Roussy, Université Paris-Saclay, UMR Radiothérapie Moléculaire, Villejuif, France. .,INSERM, U1030, SIRIC Socrates, DHU TORINO, Villejuif, France.,Gustave Roussy, Université Paris-Saclay, Département de Radiothérapie, Villejuif, France.,Univ Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
45
|
Moran-Jones K. The Therapeutic Potential of Targeting the HGF/cMET Axis in Ovarian Cancer. Mol Diagn Ther 2017; 20:199-212. [PMID: 27139908 DOI: 10.1007/s40291-016-0201-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Survival rates for ovarian cancer have remained relatively stable for the past 2 decades despite advances in surgical techniques and cytotoxic chemotherapeutics, indicating a requirement for better therapies. One pathway currently proposed for targeting is the HGF/cMET pathway. Upregulated in a number of tumour types, cMET is a tyrosine kinase receptor expressed on epithelial cells. In ovarian cancer, it has been identified as highly expressed in the four major subtypes, with expression estimates ranging from 11 to 68 % of cases. HGF, the only known ligand for cMET, is found at high levels in both serum and ascites in women with ovarian cancer, and is proposed to induce both migration and metastasis. However, clinically validated biomarkers are not yet available for either HGF or cMET, preventing a clear understanding of the true rate of overexpression, or its correlation with prognosis. Despite this, a number of agents against HGF and cMET are currently being investigated in clinical trials for multiple tumour types, including ovarian. However, a lack of patient selection, biomarker usage, and post hoc analysis correlating response with expression has resulted in the majority of these trials showing little beneficial effect from these agents, indicating that additional research is required to determine their usefulness in patients with ovarian cancer.
Collapse
Affiliation(s)
- Kim Moran-Jones
- Wolfson Wohl Cancer Research Centre, University of Glasgow, Switchback Rd, Glasgow, G61 1QH, UK. .,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria St, Sydney, NSW, 2010, Australia.
| |
Collapse
|
46
|
Progress of antibody-based inhibitors of the HGF-cMET axis in cancer therapy. Exp Mol Med 2017; 49:e307. [PMID: 28336955 PMCID: PMC5382561 DOI: 10.1038/emm.2017.17] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 12/22/2016] [Indexed: 12/27/2022] Open
Abstract
Dysregulated receptor tyrosine kinase signaling in human cancer cells leads to tumor progression, invasion and metastasis. The receptor tyrosine kinase cMET is frequently overexpressed in cancer tissue, and activation of cMET signaling is related to drug resistance and the processes of carcinogenesis, invasion and metastasis. For that reason, cMET and its ligand, hepatocyte growth factor (HGF), are considered prime targets for the development of anticancer drugs. At least eight anti-cMET and four anti-HGF antibodies have been tested or are being tested in clinical trials. However, to date none of these HGF/cMET inhibitors have shown significant efficacy in clinical trials. Furthermore, no receptor tyrosine kinase inhibitors primarily targeting cMET have been approved. Given that neutralization of HGF or cMET does not cause significant adverse effects, inhibition of the HGF/cMET signaling pathway appears to be safe. In this review, we summarized the completed and ongoing clinical trials testing antibody- or protein-based anticancer drugs targeting cMET and HGF.
Collapse
|
47
|
Cloughesy T, Finocchiaro G, Belda-Iniesta C, Recht L, Brandes AA, Pineda E, Mikkelsen T, Chinot OL, Balana C, Macdonald DR, Westphal M, Hopkins K, Weller M, Bais C, Sandmann T, Bruey JM, Koeppen H, Liu B, Verret W, Phan SC, Shames DS. Randomized, Double-Blind, Placebo-Controlled, Multicenter Phase II Study of Onartuzumab Plus Bevacizumab Versus Placebo Plus Bevacizumab in Patients With Recurrent Glioblastoma: Efficacy, Safety, and Hepatocyte Growth Factor and O 6-Methylguanine-DNA Methyltransferase Biomarker Analyses. J Clin Oncol 2017; 35:343-351. [PMID: 27918718 DOI: 10.1200/jco.2015.64.7685] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024] Open
Abstract
Purpose Bevacizumab regimens are approved for the treatment of recurrent glioblastoma in many countries. Aberrant mesenchymal-epithelial transition factor (MET) expression has been reported in glioblastoma and may contribute to bevacizumab resistance. The phase II study GO27819 investigated the monovalent MET inhibitor onartuzumab plus bevacizumab (Ona + Bev) versus placebo plus bevacizumab (Pla + Bev) in recurrent glioblastoma. Methods At first recurrence after chemoradiation, bevacizumab-naïve patients with glioblastoma were randomly assigned 1:1 to receive Ona (15 mg/kg, once every 3 weeks) + Bev (15 mg/kg, once every 3 weeks) or Pla + Bev until disease progression. The primary end point was progression-free survival by response assessment in neuro-oncology criteria. Secondary end points were overall survival, objective response rate, duration of response, and safety. Exploratory biomarker analyses correlated efficacy with expression levels of MET ligand hepatocyte growth factor, O6-methylguanine-DNA methyltransferase promoter methylation, and glioblastoma subtype. Results Among 129 patients enrolled (Ona + Bev, n = 64; Pla + Bev, n = 65), baseline characteristics were balanced. The median progression-free survival was 3.9 months for Ona + Bev versus 2.9 months for Pla + Bev (hazard ratio, 1.06; 95% CI, 0.72 to 1.56; P = .7444). The median overall survival was 8.8 months for Ona + Bev and 12.6 months for Pla + Bev (hazard ratio, 1.45; 95% CI, 0.88 to 2.37; P = .1389). Grade ≥ 3 adverse events were reported in 38.5% of patients who received Ona + Bev and 35.9% of patients who received Pla + Bev. Exploratory biomarker analyses suggested that patients with high expression of hepatocyte growth factor or unmethylated O6-methylguanine-DNA methyltransferase may benefit from Ona + Bev. Conclusion There was no evidence of further clinical benefit with the addition of onartuzumab to bevacizumab compared with bevacizumab plus placebo in unselected patients with recurrent glioblastoma in this phase II study; however, further investigation into biomarker subgroups is warranted.
Collapse
Affiliation(s)
- Timothy Cloughesy
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Gaetano Finocchiaro
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Cristóbal Belda-Iniesta
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Lawrence Recht
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Alba A Brandes
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Estela Pineda
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Tom Mikkelsen
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Olivier L Chinot
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Carmen Balana
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - David R Macdonald
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Manfred Westphal
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Kirsten Hopkins
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Michael Weller
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Carlos Bais
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Sandmann
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Jean-Marie Bruey
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Hartmut Koeppen
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Bo Liu
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - Wendy Verret
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - See-Chun Phan
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| | - David S Shames
- Timothy Cloughesy, University of California, Los Angeles; Lawrence Recht, Stanford Cancer Center, Stanford; Carlos Bais, Thomas Sandmann, Jean-Marie Bruey, Hartmut Koeppen, Bo Liu, Wendy Verret, See-Chun Phan, and David S. Shames, Genentech, South San Francisco, CA; Tom Mikkelsen, Henry Ford Hospital, Detroit, MI; Gaetano Finocchiaro, Istituto Neurologico Carlo Besta, Milan; Alba A. Brandes, Azienda Unità Sanitaria Locale-Istituto di Ricovero e Cura a Carattere Scientifico Institute of Neurologic Sciences, Bologna, Italy; Cristóbal Belda-Iniesta, Centro Integral Oncológico Clara Campal, University Hospital HM Sanchinarro, Madrid; Estela Pineda, Hospital Clinic of Barcelona; Carmen Balana, Institut Català d'Oncologia Badalona Hospital Germans Trias I Pujol, Barcelona, Spain; Olivier L. Chinot, Aix-Marseille Université Assistance Publique-Hôpitaux de Marseille, Marseille, France; David R. Macdonald, London Health Sciences Centre, London, Ontario, Canada; Manfred Westphal, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Kirsten Hopkins, Bristol Haematology and Oncology Centre, Bristol, United Kingdom; and Michael Weller, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
48
|
Imamura R, Matsumoto K. Hepatocyte growth factor in physiology and infectious diseases. Cytokine 2017; 98:97-106. [PMID: 28094206 DOI: 10.1016/j.cyto.2016.12.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/26/2016] [Accepted: 12/26/2016] [Indexed: 01/14/2023]
Abstract
Hepatocyte growth factor (HGF) is a pleiotropic cytokine composed of an α-chain and a β-chain, and these chains contain four kringle domains and a serine protease-like structure, respectively. The receptor for HGF was identified as the c-met proto-oncogene product of transmembrane receptor tyrosine kinase. HGF-induced signaling through the receptor Met provokes dynamic biological responses that support morphogenesis, regeneration, and the survival of various cells and tissues, which includes hepatocytes, renal tubular cells, and neurons. Characterization of tissue-specific Met knockout mice has further indicated that the HGF-Met system modulates immune cell functions and also plays an inhibitory role in the progression of chronic inflammation and fibrosis. However, the biological actions that are driven by the HGF-Met pathway all play a role in the acquisition of the malignant characteristics in tumor cells, such as invasion, metastasis, and drug resistance in the tumor microenvironment. Even though oncogenic Met signaling remains the major research focus, the HGF-Met axis has also been implicated in infectious diseases. Many pathogens try to utilize host HGF-Met system to establish comfortable environment for infection. Their strategies are not only simply change the expression level of HGF or Met, but also actively hijack HGF-Met system and deregulating Met signaling using their pathogenic factors. Consequently, the monitoring of HGF and Met expression, along with real-time detection of Met activation, can be a beneficial biomarker of these infectious diseases. Preclinical studies designed to address the therapeutic significance of HGF have been performed on injury/disease models, including acute tissue injury, chronic fibrosis, and cardiovascular and neurodegenerative diseases. Likewise, manipulating the HGF-Met system with complete control will lead to a tailor made treatment for those infectious diseases.
Collapse
Affiliation(s)
- Ryu Imamura
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| |
Collapse
|
49
|
Imura Y, Nakai T, Yamada S, Outani H, Takenaka S, Hamada K, Araki N, Itoh K, Yoshikawa H, Naka N. Functional and therapeutic relevance of hepatocyte growth factor/c-MET signaling in synovial sarcoma. Cancer Sci 2016; 107:1867-1876. [PMID: 27779808 PMCID: PMC5198956 DOI: 10.1111/cas.13092] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 09/26/2016] [Accepted: 09/28/2016] [Indexed: 01/02/2023] Open
Abstract
Synovial sarcoma (SS) is an aggressive soft tissue sarcoma with a poor prognosis and, thus, novel therapeutic strategies for SS are urgently required. In the present study, we investigated the functional and therapeutic relevance of hepatocyte growth factor (HGF)/c‐MET signaling in SS. Both HGF and c‐MET were highly expressed in Yamato‐SS cells, resulting in activation of c‐MET and its downstream AKT and extracellular signal‐regulated kinase signaling pathways, whereas c‐MET was expressed but not activated in SYO‐1 or HS‐SY‐II cells. c‐MET‐activated Yamato‐SS cells showed higher anchorage‐independent growth ability and less sensitivity to chemotherapeutic agents than did c‐MET‐inactivated SYO‐1 or HS‐SY‐II cells. INC280, a selective c‐MET inhibitor, inhibited growth of Yamato‐SS cells both in vitro and in vivo but not that of SYO‐1 or HS‐SY‐II cells. INC280 induced cell cycle arrest and apoptosis, and blocked phosphorylation of c‐MET and its downstream effectors in Yamato‐SS cells. Co‐expression of HGF and c‐MET in SS clinical samples correlated with a poor prognosis in patients with SS. Taken together, activation of HGF/c‐MET signaling in an autocrine fashion leads to an aggressive phenotype in SS and targeting of this signaling exerts superior antitumor effects on c‐MET‐activated SS. HGF/c‐MET expression status is a potential biomarker for identification of SS patients with a worse prognosis who can benefit from c‐MET inhibitors.
Collapse
Affiliation(s)
- Yoshinori Imura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.,Musculoskeletal Oncology Service, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Takaaki Nakai
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shutaro Yamada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hidetatsu Outani
- Musculoskeletal Oncology Service, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Satoshi Takenaka
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenichiro Hamada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Nobuhito Araki
- Musculoskeletal Oncology Service, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Kazuyuki Itoh
- Biology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norifumi Naka
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.,Biology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| |
Collapse
|
50
|
Náger M, Santacana M, Bhardwaj D, Valls J, Ferrer I, Nogués P, Cantí C, Herreros J. Nuclear phosphorylated Y142 β-catenin accumulates in astrocytomas and glioblastomas and regulates cell invasion. Cell Cycle 2016; 14:3644-55. [PMID: 26654598 DOI: 10.1080/15384101.2015.1104443] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a fast growing brain tumor characterized by extensive infiltration into the surrounding tissue and one of the most aggressive cancers. GBM is the most common glioma (originating from glial-derived cells) that either evolves from a low grade astrocytoma or appears de novo. Wnt/β-catenin and Hepatocyte Growth Factor (HGF)/c-Met signaling are hyperactive in human gliomas, where they regulate cell proliferation, migration and stem cell behavior. We previously demonstrated that β-catenin is phosphorylated at Y142 by recombinant c-Met kinase and downstream of HGF signaling in neurons. Here we studied phosphoY142 (PY142) β-catenin and dephospho S/T β-catenin (a classical Wnt transducer) in glioma biopsies, GBM cell lines and biopsy-derived glioma cell cultures. We found that PY142 β-catenin mainly localizes in the nucleus and signals through transcriptional activation in GBM cells. Tissue microarray analysis confirmed strong nuclear PY142 β-catenin immunostaining in astrocytoma and GBM biopsies. By contrast, active β-catenin showed nuclear localization only in GBM samples. Western blot analysis of tumor biopsies further indicated that PY142 and active β-catenin accumulate independently, correlating with the expression of Snail/Slug (an epithelial-mesenchymal transition marker) and Cyclin-D1 (a regulator of cell cycle progression), respectively, in high grade astrocytomas and GBMs. Moreover, GBM cells stimulated with HGF showed increasing levels of PY142 β-catenin and Snail/Slug. Importantly, the expression of mutant Y142F β-catenin decreased cell detachment and invasion induced by HGF in GBM cell lines and biopsy-derived cell cultures. Our results identify PY142 β-catenin as a nuclear β-catenin signaling form that downregulates adhesion and promotes GBM cell invasion.
Collapse
Affiliation(s)
- Mireia Náger
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| | - Maria Santacana
- b Immunohistochemical and Biostatistics and Epidemiology Units; IRBLleida ; Lleida , Spain
| | - Deepshikha Bhardwaj
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| | - Joan Valls
- b Immunohistochemical and Biostatistics and Epidemiology Units; IRBLleida ; Lleida , Spain
| | - Isidre Ferrer
- c Institute of Neuropathology; Hospital de Bellvitge-IDIBELL ; Barcelona , Spain
| | - Pere Nogués
- d Neurosurgery Unit; Hospital Arnau de Vilanova ; Lleida , Spain
| | - Carles Cantí
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| | - Judit Herreros
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| |
Collapse
|