51
|
Ding G, Wang J, Ding P, Wen Y, Yang L. Case report: HER2 amplification as a resistance mechanism to crizotinib in NSCLC with MET exon 14 skipping. Cancer Biol Ther 2019; 20:837-842. [PMID: 30744539 DOI: 10.1080/15384047.2019.1566049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Patients with non-small cell lung cancer (NSCLC) harboring MET exon 14 skipping can benefit from crizotinib treatment. Currently, the main resistance mechanisms to crizotinib are MET D1228N and Y1230C mutations. We reported a case of a Chinese NSCLC patient with MET exon 14 skipping detected by targeted next-generation sequencing (NGS) achieved clinical and imaging remission after crizotinib treatment. Then, amplification of multiple genes such as erb-b2 receptor tyrosine kinase 2 (HER2) was detected when disease progressed, indicating novel resistance mechanisms to crizotinib. Ultimately the patient died from cancer-related factors. This was the first NSCLC case with MET exon 14 skipping which reported the HER2 gene amplification at the time of progression during crizotinib treatment, indicating that bypass mechanisms contribute to the development of acquired resistance to MET inhibitors.
Collapse
Affiliation(s)
- Guanggui Ding
- a Department of Thoracic Surgery , Shenzhen People's Hospital, Second Clinical Medical College of Jinan University
| | - Jian Wang
- a Department of Thoracic Surgery , Shenzhen People's Hospital, Second Clinical Medical College of Jinan University
| | - Peikun Ding
- a Department of Thoracic Surgery , Shenzhen People's Hospital, Second Clinical Medical College of Jinan University
| | - Yuxin Wen
- a Department of Thoracic Surgery , Shenzhen People's Hospital, Second Clinical Medical College of Jinan University
| | - Lin Yang
- a Department of Thoracic Surgery , Shenzhen People's Hospital, Second Clinical Medical College of Jinan University
| |
Collapse
|
52
|
Apicella M, Giannoni E, Fiore S, Ferrari KJ, Fernández-Pérez D, Isella C, Granchi C, Minutolo F, Sottile A, Comoglio PM, Medico E, Pietrantonio F, Volante M, Pasini D, Chiarugi P, Giordano S, Corso S. Increased Lactate Secretion by Cancer Cells Sustains Non-cell-autonomous Adaptive Resistance to MET and EGFR Targeted Therapies. Cell Metab 2018; 28:848-865.e6. [PMID: 30174307 DOI: 10.1016/j.cmet.2018.08.006] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 06/06/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
The microenvironment influences cancer drug response and sustains resistance to therapies targeting receptor-tyrosine kinases. However, if and how the tumor microenvironment can be altered during treatment, contributing to resistance onset, is not known. We show that, under prolonged treatment with tyrosine kinase inhibitors (TKIs), EGFR- or MET-addicted cancer cells displayed a metabolic shift toward increased glycolysis and lactate production. We identified secreted lactate as the key molecule instructing cancer-associated fibroblasts to produce hepatocyte growth factor (HGF) in a nuclear factor κB-dependent manner. Increased HGF, activating MET-dependent signaling in cancer cells, sustained resistance to TKIs. Functional or pharmacological targeting of molecules involved in the lactate axis abrogated in vivo resistance, demonstrating the crucial role of this metabolite in the adaptive process. This adaptive resistance mechanism was observed in lung cancer patients progressed on EGFR TKIs, demonstrating the clinical relevance of our findings and opening novel scenarios in the challenge to drug resistance.
Collapse
Affiliation(s)
- Maria Apicella
- Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Torino, Candiolo 10060, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy
| | - Stephany Fiore
- Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Torino, Candiolo 10060, Italy; University of Torino, Department of Oncology, 10060, Torino, Candiolo, Italy
| | - Karin Johanna Ferrari
- Department of Experimental Oncology, European Institute of Oncology, 20139 Milan, Italy
| | | | - Claudio Isella
- Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Torino, Candiolo 10060, Italy
| | | | | | - Antonino Sottile
- Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Torino, Candiolo 10060, Italy
| | - Paolo Maria Comoglio
- Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Torino, Candiolo 10060, Italy
| | - Enzo Medico
- Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Torino, Candiolo 10060, Italy; University of Torino, Department of Oncology, 10060, Torino, Candiolo, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Marco Volante
- University of Torino, Department of Oncology, 10060, Torino, Candiolo, Italy; Pathology Unit, San Luigi Hospital, 10043 Orbassano, Italy
| | - Diego Pasini
- Department of Experimental Oncology, European Institute of Oncology, 20139 Milan, Italy; Department of Health Sciences, University of Milan, Milan, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy; Tuscany Tumor Institute and "Center for Research, Transfer and High Education DenoTHE", 50134 Florence, Italy
| | - Silvia Giordano
- Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Torino, Candiolo 10060, Italy; University of Torino, Department of Oncology, 10060, Torino, Candiolo, Italy.
| | - Simona Corso
- Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Torino, Candiolo 10060, Italy; University of Torino, Department of Oncology, 10060, Torino, Candiolo, Italy.
| |
Collapse
|
53
|
Chen S, Yang SY, Chen Z, Tan Y, Jiang YY, Chen YZ. Drug sales confirm clinical advantage of multi‐target inhibition of drug escapes by anticancer kinase inhibitors. Drug Dev Res 2018; 80:246-252. [PMID: 30422335 DOI: 10.1002/ddr.21486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/24/2018] [Accepted: 10/08/2018] [Indexed: 02/05/2023]
Affiliation(s)
- Shangying Chen
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua University Shenzhen Graduate School, Shenzhen Technology and Engineering Laboratory for Personalized Cancer Diagnostics and TherapeuticsShenzhen Kivita Innovative Drug Discovery Institute Guangdong P. R. China
| | - Sheng Yong Yang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, West China School of MedicineSichuan University Chengdu China
| | - Zhe Chen
- Zhejiang Key Laboratory of Gastro‐intestinal Pathophysiology, Zhejiang Hospital of Traditional Chinese MedicineZhejiang Chinese Medical University Hangzhou China
| | - Ying Tan
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua University Shenzhen Graduate School, Shenzhen Technology and Engineering Laboratory for Personalized Cancer Diagnostics and TherapeuticsShenzhen Kivita Innovative Drug Discovery Institute Guangdong P. R. China
| | - Yu Yang Jiang
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua University Shenzhen Graduate School, Shenzhen Technology and Engineering Laboratory for Personalized Cancer Diagnostics and TherapeuticsShenzhen Kivita Innovative Drug Discovery Institute Guangdong P. R. China
| | - Yu Zong Chen
- Bioinformatics and Drug Design Group, Department of PharmacyNational University of Singapore Singapore Singapore
| |
Collapse
|
54
|
Rashed WM. C-MET as a potential target therapy toward personalized therapy in some pediatric tumors: An overview. Crit Rev Oncol Hematol 2018; 131:7-15. [DOI: 10.1016/j.critrevonc.2018.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 08/22/2018] [Indexed: 12/28/2022] Open
|
55
|
Kim S, Kim TM, Kim DW, Kim S, Kim M, Ahn YO, Keam B, Heo DS. Acquired Resistance of MET-Amplified Non-small Cell Lung Cancer Cells to the MET Inhibitor Capmatinib. Cancer Res Treat 2018; 51:951-962. [PMID: 30309221 PMCID: PMC6639226 DOI: 10.4143/crt.2018.052] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 10/05/2018] [Indexed: 12/29/2022] Open
Abstract
Purpose Amplified mesenchymal-epithelial transition factor, MET, is a receptor tyrosine kinase (RTK) that has been considered a druggable target in non-small cell lung cancer (NSCLC). Although multiple MET tyrosine kinase inhibitors (TKIs) are being actively developed for MET-driven NSCLC, the mechanisms of acquired resistance to MET-TKIs have not been well elucidated. To understand the mechanisms of resistance and establish therapeutic strategies, we developed an in vitro model using the MET-amplified NSCLC cell line EBC-1. Materials and Methods We established capmatinib-resistant NSCLC cell lines and identified alternative signaling pathways using 3′ mRNA sequencing and human phospho-RTK arrays. Copy number alterations were evaluated by quantitative polymerase chain reaction and cell proliferation assay; activation of RTKs and downstream effectors were compared between the parental cell line EBC-1 and the resistant cell lines. Results We found that EBC-CR1 showed an epidermal growth factor receptor (EGFR)‒dependent growth and sensitivity to afatinib, an irreversible EGFR TKI. EBC-CR2 cells that had overexpression of EGFR-MET heterodimer dramatically responded to combined capmatinib with afatinib. In addition, EBC-CR3 cells derived from EBC-CR1 cells that activated EGFR with amplified phosphoinositide-3 kinase catalytic subunit α (PIK3CA) were sensitive to combined afatinib with BYL719, a phosphoinositide 3-kinase α (PI3Kα) inhibitor. Conclusion Our in vitro studies suggested that activation of EGFR signaling and/or genetic alteration of downstream effectors like PIK3CA were alternative resistance mechanisms used by capmatinib-resistant NSCLC cell lines. In addition, combined treatments with MET, EGFR, and PI3Kα inhibitors may be effective therapeutic strategies in capmatinib-resistant NSCLC patients.
Collapse
Affiliation(s)
- Seulki Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Tae Min Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dong-Wan Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Soyeon Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Miso Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yong-Oon Ahn
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Bhumsuk Keam
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dae Seog Heo
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
56
|
Virzì AR, Gentile A, Benvenuti S, Comoglio PM. Reviving oncogenic addiction to MET bypassed by BRAF (G469A) mutation. Proc Natl Acad Sci U S A 2018; 115:10058-10063. [PMID: 30224486 PMCID: PMC6176587 DOI: 10.1073/pnas.1721147115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cancer clonal evolution is based on accrual of driving genetic alterations that are expected to cooperate and progressively increase malignancy. Little is known on whether any genetic alteration can hinder the oncogenic function of a coexisting alteration, so that therapeutic targeting of the one can, paradoxically, revive the function of the other. We report the case of a driver oncogene (MET) that is not only bypassed, but also disabled by the mutation of a downstream transducer (BRAF), and reignited by inhibition of the latter. In a metastasis originated from a cancer of unknown primary (CUP), the MET oncogene was amplified eightfold, but unexpectedly, the kinase was dephosphorylated and inactive. As result, specific drugs targeting MET (JNJ-38877605) failed to inhibit growth of xenografts derived from the patient. In addition to MET amplification, the patient harbored, as sole proliferative driver, a mutation hyperactivating BRAF (G469A). Surprisingly, specific blockade of the BRAF pathway was equally ineffective, and it was accompanied by rephosphorylation of the amplified MET oncoprotein and by revived addiction to MET. Mechanistically, MET inactivation in the context of the BRAF-activating mutation is driven through a negative feedback loop involving inactivation of PP2A phosphatase, which in turn leads to phosphorylation on MET inhibitory Ser985. Disruption of this feedback loop allows PP2A reactivation, removing the inhibitory phosphorylation from Ser985 and thereby unleashing MET kinase activity. Evidence is provided for a mechanism of therapeutic resistance to single-oncoprotein targeting, based on reactivation of a genetic alteration functionally dormant in targeted cancer cells.
Collapse
Affiliation(s)
- Anna Rita Virzì
- Laboratory of Molecular Therapeutics and Exploratory Research, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia- Istituto di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), 10060 Candiolo, Italy
| | - Alessandra Gentile
- Laboratory of Molecular Therapeutics and Exploratory Research, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia- Istituto di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), 10060 Candiolo, Italy
| | - Silvia Benvenuti
- Laboratory of Molecular Therapeutics and Exploratory Research, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia- Istituto di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), 10060 Candiolo, Italy
| | - Paolo M Comoglio
- Laboratory of Molecular Therapeutics and Exploratory Research, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia- Istituto di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), 10060 Candiolo, Italy
| |
Collapse
|
57
|
Baldacci S, Kherrouche Z, Descarpentries C, Wislez M, Dansin E, Furlan A, Tulasne D, Cortot AB. [MET exon 14 splicing sites mutations: A new therapeutic opportunity in lung cancer]. Rev Mal Respir 2018; 35:796-812. [PMID: 30174236 DOI: 10.1016/j.rmr.2018.01.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 01/08/2018] [Indexed: 01/23/2023]
Abstract
The mutations leading to MET exon 14 skipping represent a new class of molecular alterations described in various cancers. These alterations are observed in 2 to 3 % of cases of non-small cell lung cancer (NSCLC). Several cases of NSCLC carrying such alterations and achieving objective response to MET tyrosine kinase inhibitorshave recently been published. This review summarizes the molecular mechanisms responsible for MET exon 14 skipping as well as the consequences of the loss of this exon on receptor activity. We also describe the clinical characteristics of patients with METΔ14 mutations. Finally, we address the issues related to the detection of these mutations in lung cancer, and the need to anticipate resistance to MET inhibitors.
Collapse
Affiliation(s)
- S Baldacci
- Université Lille, CNRS, UMR 8161-M3T, mécanismes de tumorigenèse et thérapies ciblées, Institut Pasteur de Lille, 59000 Lille, France; Service de pneumologie et oncologie thoracique, OncoLille, université de Lille, CHU de Lille, 59000 Lille, France
| | - Z Kherrouche
- Université Lille, CNRS, UMR 8161-M3T, mécanismes de tumorigenèse et thérapies ciblées, Institut Pasteur de Lille, 59000 Lille, France
| | - C Descarpentries
- Service de biochimie et biologie moléculaire, plateforme de biologie moléculaire des cancers, CHU de Lille, 59000 Lille, France
| | - M Wislez
- Service de pneumologie, hôpital Tenon, AP-HP, 75020 Paris , France
| | - E Dansin
- Département de cancérologie cervico-faciale & thoracique, CLCC Oscar Lambret, 59000 Lille, France
| | - A Furlan
- Université Lille, CNRS, UMR 8161-M3T, mécanismes de tumorigenèse et thérapies ciblées, Institut Pasteur de Lille, 59000 Lille, France; Équipe de biophotonique cellulaire fonctionnelle, université Lille, CNRS UMR 8523 PhLAM, 59000 Lille, France
| | - D Tulasne
- Université Lille, CNRS, UMR 8161-M3T, mécanismes de tumorigenèse et thérapies ciblées, Institut Pasteur de Lille, 59000 Lille, France
| | - A B Cortot
- Université Lille, CNRS, UMR 8161-M3T, mécanismes de tumorigenèse et thérapies ciblées, Institut Pasteur de Lille, 59000 Lille, France; Service de pneumologie et oncologie thoracique, OncoLille, université de Lille, CHU de Lille, 59000 Lille, France.
| |
Collapse
|
58
|
Miranda O, Farooqui M, Siegfried JM. Status of Agents Targeting the HGF/c-Met Axis in Lung Cancer. Cancers (Basel) 2018; 10:cancers10090280. [PMID: 30134579 PMCID: PMC6162713 DOI: 10.3390/cancers10090280] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 12/15/2022] Open
Abstract
Hepatocyte growth factor (HGF) is the ligand for the tyrosine kinase receptor c-Met (Mesenchymal Epithelial Transition Factor also known as Hepatocyte Growth Factor Receptor, HGFR), a receptor with expression throughout epithelial and endothelial cell types. Activation of c-Met enhances cell proliferation, invasion, survival, angiogenesis, and motility. The c-Met pathway also stimulates tissue repair in normal cells. A body of past research shows that increased levels of HGF and/or overexpression of c-Met are associated with poor prognosis in several solid tumors, including lung cancer, as well as cancers of the head and neck, gastro-intestinal tract, breast, ovary and cervix. The HGF/c-Met signaling network is complex; both ligand-dependent and ligand-independent signaling occur. This article will provide an update on signaling through the HGF/c-Met axis, the mechanism of action of HGF/c-Met inhibitors, the lung cancer patient populations most likely to benefit, and possible mechanisms of resistance to these inhibitors. Although c-Met as a target in non-small cell lung cancer (NSCLC) showed promise based on preclinical data, clinical responses in NSCLC patients have been disappointing in the absence of MET mutation or MET gene amplification. New therapeutics that selectively target c-Met or HGF, or that target c-Met and a wider spectrum of interacting tyrosine kinases, will be discussed.
Collapse
Affiliation(s)
- Oshin Miranda
- Department of Pharmacology and Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Mariya Farooqui
- Department of Pharmacology and Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jill M Siegfried
- Department of Pharmacology and Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
- Department of Pharmacology, University of Minnesota, 321 Church Street SE, 6-120 Jackson Hall, Minneapolis, MN 55455, USA.
| |
Collapse
|
59
|
Baro M, Lopez Sambrooks C, Quijano A, Saltzman WM, Contessa J. Oligosaccharyltransferase Inhibition Reduces Receptor Tyrosine Kinase Activation and Enhances Glioma Radiosensitivity. Clin Cancer Res 2018; 25:784-795. [PMID: 29967251 DOI: 10.1158/1078-0432.ccr-18-0792] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/21/2018] [Accepted: 06/27/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Parallel signaling reduces the effects of receptor tyrosine kinase (RTK)-targeted therapies in glioma. We hypothesized that inhibition of protein N-linked glycosylation, an endoplasmic reticulum co- and posttranslational modification crucial for RTK maturation and activation, could provide a new therapeutic approach for glioma radiosensitization.Experimental Design: We investigated the effects of a small-molecule inhibitor of the oligosaccharyltransferase (NGI-1) on EGFR family receptors, MET, PDGFR, and FGFR1. The influence of glycosylation state on tumor cell radiosensitivity, chemotherapy-induced cell toxicity, DNA damage, and cell-cycle arrest were determined and correlated with glioma cell receptor expression profiles. The effects of NGI-1 on xenograft tumor growth were tested using a nanoparticle formulation validated by in vivo molecular imaging. A mechanistic role for RTK signaling was evaluated through the expression of a glycosylation-independent CD8-EGFR chimera. RESULTS NGI-1 reduced glycosylation, protein levels, and activation of most RTKs. NGI-1 also enhanced the radiosensitivity and cytotoxic effects of chemotherapy in those glioma cells with elevated ErbB family activation, but not in cells without high levels of RTK activation. NGI-1 radiosensitization was associated with increases in both DNA damage and G1 cell-cycle arrest. Combined treatment of glioma xenografts with fractionated radiotherapy and NGI-1 significantly reduced tumor growth compared with controls. Expression of the CD8-EGFR eliminated the effects of NGI-1 on G1 arrest, DNA damage, and cellular radiosensitivity, identifying RTK inhibition as the principal mechanism for the NGI-1 effect. CONCLUSIONS This study suggests that oligosaccharyltransferase inhibition with NGI-1 is a novel approach to radiosensitize malignant gliomas with enhanced RTK signaling.See related commentary by Wahl and Lawrence, p. 455.
Collapse
Affiliation(s)
- Marta Baro
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut
| | | | - Amanda Quijano
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Joseph Contessa
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut. .,Department of Pharmacology, Yale University, New Haven, Connecticut
| |
Collapse
|
60
|
Konstantinopoulos PA, Brady WE, Farley J, Armstrong A, Uyar DS, Gershenson DM. Phase II study of single-agent cabozantinib in patients with recurrent clear cell ovarian, primary peritoneal or fallopian tube cancer (NRG-GY001). Gynecol Oncol 2018; 150:9-13. [PMID: 29739622 PMCID: PMC6365003 DOI: 10.1016/j.ygyno.2018.04.572] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/28/2018] [Accepted: 04/30/2018] [Indexed: 10/17/2022]
Abstract
OBJECTIVE To evaluate the efficacy and tolerability of cabozantinib in recurrent clear cell ovarian, primary peritoneal or fallopian tube cancer. METHODS Patients with recurrent ovarian, fallopian or primary peritoneal tumors with at least 50% clear cell histomorphology, measurable disease, one or two prior regimens and ECOG performance status 0-2 received cabozantinib 60 mg orally once daily continuously, in 4-week cycles until disease progression or unacceptable toxicity. Primary endpoints were progression-free survival (PFS) at six months and complete or partial tumor response (as assessed by RECIST 1.1). Secondary endpoints included toxicity, PFS, and overall survival (OS). RESULTS Over 19 months, 13 patients were accrued. Fifty-four percent of patients were ≥60 years of age. Performance statuses of 0 and 1 comprised 8 and 5 patients. No objective tumor responses were seen. Three (23% [95% CI: 5%, 54%]) of 13 patients had PFS ≥6 months, including one patient who received cabozantinib for 23 cycles and was still on treatment as of the data cut-off date. Median PFS and OS were 3.6 and 8.1 months, respectively. There was one patient with a grade 5 event: a thromboembolic event considered possibly related to study therapy; patient's cause of death was determined to be due to disease and protocol treatment. Four other patients had thromboembolic events (two grade 3 and one each grade 1 and grade 2). Other grade 3 or higher events reported in two or more patients were nausea, vomiting, fatigue, dyspnea, and dehydration. CONCLUSIONS Cabozantinib demonstrated minimal activity in the second- and third-line treatments of clear cell ovarian, fallopian tube or primary peritoneal carcinoma.
Collapse
Affiliation(s)
| | - William E Brady
- NRG Oncology, Clinical Trial Development Division, Biostatistics & Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, United States.
| | - John Farley
- Department of Obstetrics & Gynecology, Uniformed Serviced University of the Health Sciences, Bethesda, MD, United States.
| | - Amy Armstrong
- Obstetrics and Gynecology, CWRU School of Medicine, Cleveland, OH, United States.
| | - Denise S Uyar
- Gynecologic Oncology, Medical School of Wisconsin, Milwaukee, WI 53226, United States.
| | - David M Gershenson
- Department of Gynecologic Oncology & Reproductive Medicine, Unit 1362, The University of Texas, MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
61
|
Shin SY, Müller AK, Verma N, Lev S, Nguyen LK. Systems modelling of the EGFR-PYK2-c-Met interaction network predicts and prioritizes synergistic drug combinations for triple-negative breast cancer. PLoS Comput Biol 2018; 14:e1006192. [PMID: 29920512 PMCID: PMC6007894 DOI: 10.1371/journal.pcbi.1006192] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 05/10/2018] [Indexed: 12/18/2022] Open
Abstract
Prediction of drug combinations that effectively target cancer cells is a critical challenge for cancer therapy, in particular for triple-negative breast cancer (TNBC), a highly aggressive breast cancer subtype with no effective targeted treatment. As signalling pathway networks critically control cancer cell behaviour, analysis of signalling network activity and crosstalk can help predict potent drug combinations and rational stratification of patients, thus bringing therapeutic and prognostic values. We have previously showed that the non-receptor tyrosine kinase PYK2 is a downstream effector of EGFR and c-Met and demonstrated their crosstalk signalling in basal-like TNBC. Here we applied a systems modelling approach and developed a mechanistic model of the integrated EGFR-PYK2-c-Met signalling network to identify and prioritize potent drug combinations for TNBC. Model predictions validated by experimental data revealed that among six potential combinations of drug pairs targeting the central nodes of the network, including EGFR, c-Met, PYK2 and STAT3, co-targeting of EGFR and PYK2 and to a lesser extent of EGFR and c-Met yielded strongest synergistic effect. Importantly, the synergy in co-targeting EGFR and PYK2 was linked to switch-like cell proliferation-associated responses. Moreover, simulations of patient-specific models using public gene expression data of TNBC patients led to predictive stratification of patients into subgroups displaying distinct susceptibility to specific drug combinations. These results suggest that mechanistic systems modelling is a powerful approach for the rational design, prediction and prioritization of potent combination therapies for individual patients, thus providing a concrete step towards personalized treatment for TNBC and other tumour types. We applied a systems modelling approach combining mechanistic modelling and biological experimentation to identify effective drug combinations for triple-negative breast cancer (TNBC), an aggressive subtype of breast cancer with no approved targeted treatment. The model predicted and prioritized the synergistic combinations as confirmed by experimental data, demonstrating the power of this approach. Moreover, analysis of clinical data of TNBC patients and patient-specific modelling simulation enabled us to stratify the patients into subgroups with distinct susceptibility to specific drug combinations, and thus defined a subset of patient that could benefit from the combined treatments.
Collapse
Affiliation(s)
- Sung-Young Shin
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | | | - Nandini Verma
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
- * E-mail: (SL); (LKN)
| | - Lan K. Nguyen
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- * E-mail: (SL); (LKN)
| |
Collapse
|
62
|
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: 1.0] [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
|
63
|
Anestis A, Zoi I, Karamouzis MV. Current advances of targeting HGF/c-Met pathway in gastric cancer. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:247. [PMID: 30069449 PMCID: PMC6046293 DOI: 10.21037/atm.2018.04.42] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 04/18/2018] [Indexed: 12/18/2022]
Abstract
Despite the advances in systemic chemotherapy, gastric adenocarcinoma (GC) remains the third most common cause of cancer-related deaths with poor prognosis. The heterogeneity of GC indicates that novel biomarkers should be established in order to further classify tumors and develop individual targeted therapies. High-quality preclinical and clinical research has demonstrated that growth factor (HGF)-hepatocyte growth factor receptor (c-Met) pathway plays a pivotal role on the growth, survival and invasiveness of GC. In particular, aberrant activation of HGF/c-Met signaling pathway has been associated with poor clinical outcomes, suggesting the therapeutic potential of c-Met. This has stimulated the development and evaluation of a number of c-Met targeted agents in an advance disease setting. In this review, we summarize the current state of the art in the advances on the inhibition of c-Met pathway, with particular emphasis on the clinical testing of c-Met targeted therapeutic agents. Furthermore, we discuss the challenges facing the incorporation of c-Met targeted agents in randomized trials, with the idea that the definition of the appropriate genetic and molecular context for the use of these agents remains the priority.
Collapse
Affiliation(s)
- Aristomenis Anestis
- Molecular Oncology Unit, Department of Biological Chemistry, ‘Laiko’ General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ilianna Zoi
- Molecular Oncology Unit, Department of Biological Chemistry, ‘Laiko’ General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Michalis V. Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, ‘Laiko’ General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Internal Medicine, ‘Laiko’ General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
64
|
Combinatorial inhibition of PTPN12-regulated receptors leads to a broadly effective therapeutic strategy in triple-negative breast cancer. Nat Med 2018; 24:505-511. [PMID: 29578538 DOI: 10.1038/nm.4507] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/29/2018] [Indexed: 12/28/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer diagnosed in more than 200,000 women each year and is recalcitrant to targeted therapies. Although TNBCs harbor multiple hyperactive receptor tyrosine kinases (RTKs), RTK inhibitors have been largely ineffective in TNBC patients thus far. We developed a broadly effective therapeutic strategy for TNBC that is based on combined inhibition of receptors that share the negative regulator PTPN12. Previously, we and others identified the tyrosine phosphatase PTPN12 as a tumor suppressor that is frequently inactivated in TNBC. PTPN12 restrains several RTKs, suggesting that PTPN12 deficiency leads to aberrant activation of multiple RTKs and a co-dependency on these receptors. This in turn leads to the therapeutic hypothesis that PTPN12-deficient TNBCs may be responsive to combined RTK inhibition. However, the repertoire of RTKs that are restrained by PTPN12 in human cells has not been systematically explored. By methodically identifying the suite of RTK substrates (MET, PDGFRβ, EGFR, and others) inhibited by PTPN12, we rationalized a combination RTK-inhibitor therapy that induced potent tumor regression across heterogeneous models of TNBC. Orthogonal approaches revealed that PTPN12 was recruited to and inhibited these receptors after ligand stimulation, thereby serving as a feedback mechanism to limit receptor signaling. Cancer-associated mutation of PTPN12 or reduced PTPN12 protein levels diminished this feedback mechanism, leading to aberrant activity of these receptors. Restoring PTPN12 protein levels restrained signaling from RTKs, including PDGFRβ and MET, and impaired TNBC survival. In contrast with single agents, combined inhibitors targeting the PDGFRβ and MET receptors induced the apoptosis in TNBC cells in vitro and in vivo. This therapeutic strategy resulted in tumor regressions in chemo-refractory patient-derived TNBC models. Notably, response correlated with PTPN12 deficiency, suggesting that impaired receptor feedback may establish a combined addiction to these proto-oncogenic receptors. Taken together, our data provide a rationale for combining RTK inhibitors in TNBC and other malignancies that lack receptor-activating mutations.
Collapse
|
65
|
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: 5.0] [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
|
66
|
Pollmann SE, Calvert VS, Rao S, Boca SM, Madhavan S, Horak ID, Kjaer A, Petricoin EF, Kragh M, Poulsen TT. Acquired Resistance to a MET Antibody In Vivo Can Be Overcome by the MET Antibody Mixture Sym015. Mol Cancer Ther 2018; 17:1259-1270. [DOI: 10.1158/1535-7163.mct-17-0787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/08/2018] [Accepted: 03/07/2018] [Indexed: 11/16/2022]
|
67
|
Acquired savolitinib resistance in non-small cell lung cancer arises via multiple mechanisms that converge on MET-independent mTOR and MYC activation. Oncotarget 2018; 7:57651-57670. [PMID: 27472392 PMCID: PMC5295379 DOI: 10.18632/oncotarget.10859] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 07/13/2016] [Indexed: 12/15/2022] Open
Abstract
Lung cancer is the most common cause of cancer death globally with a significant, unmet need for more efficacious treatments. The receptor tyrosine kinase MET has been implicated as an oncogene in numerous cancer subtypes, including non-small cell lung cancer (NSCLC). Here we explore the therapeutic potential of savolitinib (volitinib, AZD6094, HMPL-504), a potent and selective MET inhibitor, in NSCLC. In vitro, savolitinib inhibits MET phosphorylation with nanomolar potency, which correlates with blockade of PI3K/AKT and MAPK signaling as well as MYC down-regulation. In vivo, savolitinib causes inhibition of these pathways and significantly decreases growth of MET-dependent xenografts. To understand resistance mechanisms, we generated savolitinib resistance in MET-amplified NSCLC cell lines and analyzed individual clones. We found that upregulation of MYC and constitutive mTOR pathway activation is a conserved feature of resistant clones that can be overcome by knockdown of MYC or dual mTORC1/2 inhibition. Lastly, we demonstrate that mechanisms of resistance are heterogeneous, arising via a switch to EGFR dependence or by a requirement for PIM signaling. This work demonstrates the efficacy of savolitinib in NSCLC and characterizes acquired resistance, identifying both known and novel mechanisms that may inform combination strategies in the clinic.
Collapse
|
68
|
Bcl-2 and Bcl-xL mediate resistance to receptor tyrosine kinase-targeted therapy in lung and gastric cancer. Anticancer Drugs 2018; 28:1141-1149. [PMID: 28885267 DOI: 10.1097/cad.0000000000000561] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Promising clinical efficacy has been observed with receptor tyrosine kinase inhibitors (TKIs) particularly in lung and gastric cancers with mutations or amplifications in the targeted receptor tyrosine kinases (RTKs). However, the efficacy and the duration of the response to these inhibitors are limited by the emergence of drug resistance. Here, we report treatment of RTK-dependent lung and gastric cancer cell lines with TKIs increased protein levels of Bcl-2 and Bcl-xL. The combination of the Bcl-2 and Bcl-xL inhibitor ABT-263 and TKIs was superior to TKIs alone in reducing cell viability and capacity of resistant colony formation. Furthermore, resistant cells established with exposure of RTK-dependent cells to increasing concentrations of TKIs also express higher levels of Bcl-2 or Bcl-xL compared with their parental cells. The combination of inhibitors of PI3K/AKT, MEK/ERK, and Bcl-2/Bcl-xL effectively reduced the viability of resistant cells and inhibited tumor size in a xenograft model derived from resistant cells by inducing apoptosis. Our results define a generalizable resistance mechanism to TKIs and rationalize inhibition of Bcl-2 and Bcl-xL as a strategy to augment responses and blunt acquired resistance to TKIs in lung and gastric cancer.
Collapse
|
69
|
The Prognostic Significance of c-MET and EGFR Overexpression in Resected Gastric Adenocarcinomas. Am J Clin Oncol 2017; 40:543-551. [PMID: 26125303 DOI: 10.1097/coc.0000000000000202] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Epidermal growth factor receptor (EGFR) and c-MET are tyrosine kinase growth factor receptors implicated in gastric cancer (GC), and their pathways appear to be interdependent. The aim of this study was to investigate the prognostic value of EGFR and c-MET protein overexpression by immunohistochemistry in Canadian patients with resected GC and correlate it with clinicopathologic characteristics and overall survival (OS). MATERIALS AND METHODS Tissue microarray blocks were constructed from 120 resected GCs stained with EGFR and c-MET and scored semiquantitatively (0 to 3+). Each receptor's expression was compared with clinicopathologic characteristics and survival. Descriptive statistics, Kaplan-Meyer, and Cox regression were used for statistical analyses. RESULTS Of the 113 interpretable cases, overexpression of EGFR and c-MET was noted in 17 (15%) and 65 (57%), respectively; coexpression of EGFR and c-MET was observed in 12 (10%) of GC. EGFR and c-MET overexpression correlated with poor OS: median 13 versus 30 months in EGFR positive versus negative GC (hazard ratio [HR]=1.67, P=0.11); 27 versus 49 months in c-MET positive versus negative GC (HR=1.17, P=0.49), respectively. GC coexpressing EGFR and c-MET was significantly correlated with poor survival: 12 versus 29 months in double-positive versus rest of tumors both in univariate (HR=2.62, P=0.003) and multivariate analyses (HR=2.58, P=0.01). CONCLUSIONS This study describes the prevalence and prognostic value of EGFR and c-MET in a Canadian population of patients undergoing curative intent resection for GC. Both c-MET and EGFR overexpression trended toward poor OS, but only the group with EGFR+/c-MET+ GC reached statistical significance on multivariate analysis.
Collapse
|
70
|
Bahrami A, Shahidsales S, Khazaei M, Ghayour-Mobarhan M, Maftouh M, Hassanian SM, Avan A. C-Met as a potential target for the treatment of gastrointestinal cancer: Current status and future perspectives. J Cell Physiol 2017; 232:2657-2673. [PMID: 28075018 DOI: 10.1002/jcp.25794] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 01/05/2025]
Abstract
Aberrant activation of the HGF/c-Met signalling pathways is shown to be related with cell proliferation, progression, metastasis, and worse prognosis in several tumor types, including gastrointestinal cancers, suggesting its value as a stimulating-target for cancer-therapy. Several approaches have been developed for targeting HGF and/or c-Met, and one of them, crizotinib (dual c-Met/ALK inhibitor), is recently been approved by FDA for lung-cancers with ALK-rearrangement. The main aim of current review is to give an overview on the role of c-Met/HGF pathway in gastrointestinal cancer, in preclinical and clinical trials. Although several important matters is still remained to be elucidated on the molecular pathways underlying the antitumor effects of this therapy in gastrointestinal-cancers. Further investigations are warranted to recognize the main determinants of the activity of c-Met inhibitors, for parallel targeting signalling pathway associated/activated via MET/HGF pathway or in response to the cell resistance to anti-c-Met agents. Additionally, identification of patients that might benefit from therapy could help to increase the selectivity and efficacy of the therapy.
Collapse
Affiliation(s)
- Afsane Bahrami
- Molecular Medicine Group, Department of Modern Sciences and Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodabeh Shahidsales
- Cancer Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Neurogenic Inflammatory Research Center and Department of Physiology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftouh
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
71
|
Engstrom LD, Aranda R, Lee M, Tovar EA, Essenburg CJ, Madaj Z, Chiang H, Briere D, Hallin J, Lopez-Casas PP, Baños N, Menendez C, Hidalgo M, Tassell V, Chao R, Chudova DI, Lanman RB, Olson P, Bazhenova L, Patel SP, Graveel C, Nishino M, Shapiro GI, Peled N, Awad MM, Jänne PA, Christensen JG. Glesatinib Exhibits Antitumor Activity in Lung Cancer Models and Patients Harboring MET Exon 14 Mutations and Overcomes Mutation-mediated Resistance to Type I MET Inhibitors in Nonclinical Models. Clin Cancer Res 2017; 23:6661-6672. [DOI: 10.1158/1078-0432.ccr-17-1192] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/13/2017] [Accepted: 07/24/2017] [Indexed: 11/16/2022]
|
72
|
Oddo D, Siravegna G, Gloghini A, Vernieri C, Mussolin B, Morano F, Crisafulli G, Berenato R, Corti G, Volpi CC, Buscarino M, Niger M, Dunne PD, Rospo G, Valtorta E, Bartolini A, Fucà G, Lamba S, Martinetti A, Di Bartolomeo M, de Braud F, Bardelli A, Pietrantonio F, Di Nicolantonio F. Emergence of MET hyper-amplification at progression to MET and BRAF inhibition in colorectal cancer. Br J Cancer 2017; 117:347-352. [PMID: 28654634 PMCID: PMC5537500 DOI: 10.1038/bjc.2017.196] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/31/2017] [Accepted: 06/01/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Combined MET and BRAF inhibition showed clinical benefit in a patient with rectal cancer carrying BRAFV600E and MET amplification. However after 4 months, acquired resistance emerged and the patient deceased shortly after disease progression. The mechanism of resistance to this drug combination is unknown. METHODS We analysed plasma circulating tumour DNA obtained at progression by exome sequencing and digital PCR. MET gene and mRNA in situ hybridisation analyses in two bioptic specimens obtained at progression were used to confirm the plasma data. RESULTS We identified in plasma MET gene hyper-amplification as a potential mechanism underlying therapy resistance. Increased MET gene copy and transcript levels were detected in liver and lymph node metastatic biopsies. Finally, transduction of MET in BRAF mutant colorectal cancer cells conferred refractoriness to BRAF and MET inhibition. CONCLUSIONS We identified in a rectal cancer patient MET gene hyper-amplification as mechanism of resistance to dual BRAF and MET inhibition.
Collapse
Affiliation(s)
- Daniele Oddo
- Department of Oncology, University of Torino, Candiolo (TO) 10060, Italy
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO) 10060, Italy
| | - Giulia Siravegna
- Department of Oncology, University of Torino, Candiolo (TO) 10060, Italy
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO) 10060, Italy
- FIRC Institute of Molecular Oncology (IFOM), Milan 20139, Italy
| | - Annunziata Gloghini
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | | | - Federica Morano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Giovanni Crisafulli
- Department of Oncology, University of Torino, Candiolo (TO) 10060, Italy
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO) 10060, Italy
| | - Rosa Berenato
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Giorgio Corti
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO) 10060, Italy
| | - Chiara Costanza Volpi
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | | | - Monica Niger
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Philip D Dunne
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Giuseppe Rospo
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO) 10060, Italy
| | - Emanuele Valtorta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan 20162, Italy
| | - Alice Bartolini
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO) 10060, Italy
| | - Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Simona Lamba
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO) 10060, Italy
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Maria Di Bartolomeo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
- Department of Oncology, Università degli Studi di Milano, Milan 20122, Italy
| | - Alberto Bardelli
- Department of Oncology, University of Torino, Candiolo (TO) 10060, Italy
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO) 10060, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Federica Di Nicolantonio
- Department of Oncology, University of Torino, Candiolo (TO) 10060, Italy
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO) 10060, Italy
| |
Collapse
|
73
|
Chan XY, Singh A, Osman N, Piva TJ. Role Played by Signalling Pathways in Overcoming BRAF Inhibitor Resistance in Melanoma. Int J Mol Sci 2017; 18:ijms18071527. [PMID: 28708099 PMCID: PMC5536016 DOI: 10.3390/ijms18071527] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/06/2017] [Accepted: 07/12/2017] [Indexed: 12/15/2022] Open
Abstract
The discovery of the BRAFV600E mutation led to the development of vemurafenib (PLX4032), a selective BRAF inhibitor specific to the kinase, for the treatment of metastatic melanomas. However, initial success of the drug was dampened by the development of acquired resistance. Melanoma was shown to relapse in patients following treatment with vemurafenib which eventually led to patients' deaths. It has been proposed that mechanisms of resistance can be due to (1) reactivation of the mitogen-activated protein kinase (MAPK) signalling pathway via secondary mutations, amplification or activation of target kinase(s), (2) the bypass of oncogenic pathway via activation of alternative signalling pathways, (3) other uncharacterized mechanisms. Studies showed that receptor tyrosine kinases (RTK) such as PDGFRβ, IGF1R, EGFR and c-Met were overexpressed in melanoma cells. Along with increased secretion of growth factors such as HGF and TGF-α, this will trigger intracellular signalling cascades. This review discusses the role MAPK and Phosphatidylinositol-3-kinase-protein kinase B-mammalian target of rapamycin (PI3K-AKT-mTOR) pathways play in the mechanism of resistance of melanomas.
Collapse
Affiliation(s)
- Xian Yang Chan
- School of Health & Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| | - Alamdeep Singh
- School of Health & Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| | - Narin Osman
- School of Health & Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
- Department of Immunology, Monash University, Melbourne 3004, Victoria, Australia.
- Department of Pharmacy, University of Queensland, Woolloongabba 4102, Queensland, Australia.
| | - Terrence J Piva
- School of Health & Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| |
Collapse
|
74
|
Effect of AXL on the epithelial-to-mesenchymal transition in non-small cell lung cancer. Exp Ther Med 2017; 14:785-790. [PMID: 28673000 DOI: 10.3892/etm.2017.4532] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 01/20/2016] [Indexed: 01/06/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the leading cause of cancer-associated mortality in the United States. AXL, which is a member of the receptor tyrosine kinases, has been established as a strong candidate for the targeted therapy of cancer. Therefore, the present study aimed to investigate the role of AXL in NSCLC; in particular the molecular mechanisms underlying the involvement of AXL in the epithelial-to-mesenchymal transition (EMT). Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis demonstrated that AXL, EMT-inducing Twist and the mesenchymal marker N-cadherin were upregulated, and the epithelial markers E-cadherin and β-cadherin were downregulated, in the PC9 NSCLC cell line. Furthermore, downregulation of AXL expression by RNA interference was shown to inhibit cell growth by inducing the apoptosis of PC9 cells, as demonstrated by MTT and flow cytometry analyses. Notably, inhibition of AXL attenuated the regulation of EMT-associated genes, specifically downregulating Twist and N-cadherin, and upregulating E-cadherin and β-cadherin. Conversely, downregulation of Twist did not affect the expression levels of AXL. These results suggested that AXL may inhibit the EMT by the regulation of EMT-associated genes in the PC9 cell line. The results of the present study indicated that AXL may have a role in the regulation of EMT and the cell cycle of the PC9 cells; thus suggesting that AXL may have clinical significance in the design of therapeutic strategies targeting NSCLC and EMT signaling pathways.
Collapse
|
75
|
Hu CT, Wu JR, Cheng CC, Wu WS. The Therapeutic Targeting of HGF/c-Met Signaling in Hepatocellular Carcinoma: Alternative Approaches. Cancers (Basel) 2017; 9:cancers9060058. [PMID: 28587113 PMCID: PMC5483877 DOI: 10.3390/cancers9060058] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/23/2017] [Accepted: 05/24/2017] [Indexed: 12/22/2022] Open
Abstract
The poor prognosis of hepatocellular carcinoma (HCC), one of the most devastating cancers worldwide, is due to frequent recurrence and metastasis. Among the metastatic factors in the tumor microenvironment, hepatocyte growth factor (HGF) has been well known to play critical roles in tumor progression, including HCC. Therefore, c-Met is now regarded as the most promising therapeutic target for the treatment of HCC. However, there are still concerns about resistance and the side effects of using conventional inhibitors of c-Met, such as tyrosine kinase inhibitors. Recently, many alternative strategies of c-Met targeting have been emerging. These include targeting the downstream effectors of c-Met, such as hydrogen peroxide-inducible clone 5 (Hic-5), to block the reactive oxygen species (ROS)-mediated signaling for HCC progression. Also, inhibition of endosomal regulators, such as PKCε and GGA3, may perturb the c-Met endosomal signaling for HCC cell migration. On the other hand, many herbal antagonists of c-Met-dependent signaling, such as saponin, resveratrol, and LZ-8, were identified. Taken together, it can be anticipated that more effective and safer c-Met targeting strategies for preventing HCC progression can be established in the future.
Collapse
Affiliation(s)
- Chi-Tan Hu
- Research Centre for Hepatology, Department of Internal Medicine, Buddhist Tzu Chi General Hospital and Tzu Chi University, Hualien 970, Taiwan.
| | - Jia-Ru Wu
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Chuan-Chu Cheng
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Wen-Sheng Wu
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| |
Collapse
|
76
|
Li A, Yang JJ, Zhang XC, Zhang Z, Su J, Gou LY, Bai Y, Zhou Q, Yang Z, Han-Zhang H, Zhong WZ, Chuai S, Zhang Q, Xie Z, Gao H, Chen H, Wang Z, Wang Z, Yang XN, Wang BC, Gan B, Chen ZH, Jiang BY, Wu SP, Liu SY, Xu CR, Wu YL. Acquired MET Y1248H and D1246N Mutations Mediate Resistance to MET Inhibitors in Non-Small Cell Lung Cancer. Clin Cancer Res 2017; 23:4929-4937. [PMID: 28396313 DOI: 10.1158/1078-0432.ccr-16-3273] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 12/20/2016] [Accepted: 04/04/2017] [Indexed: 11/16/2022]
Abstract
Purpose:MET amplification, responsible for 20% of acquired resistance to EGFR tyrosine kinase inhibitor (TKI) in patients with advanced non-small cell lung cancer (NSCLC), presents an attractive target. Numerous studies have conferred susceptibility of MET mutations and focal amplification to targeted MET-TKIs. However, the mechanism underlying MET-TKIs-induced resistance remains elusive.Experimental Design: We conducted a cohort of 12 patients with advanced NSCLC who developed resistance to a combinatorial therapy consisting of gefitinib and a type I MET-TKI. We performed capture-based targeted ultra-deep sequencing on serial tumor biopsies and plasmas ctDNA samples to detect and quantify genetic alterations.Results: We identified 2 newly acquired MET mutations, Y1248H and D1246N, in 2 patients and further confirmed their resistance against type I MET-TKIs in silco, in vitro, and in vivo Interestingly, NIH3T3 cells harboring either mutation exhibited responses to type II MET-TKIs, suggesting sequential use of MET-TKIs may offer a more durable response. In addition, we also discovered that EGFR amplification may act as an alternative MET-TKI resistance mechanism.Conclusions: Our study provides insight into the diversity of mechanisms underlying MET-TKI-induced resistance and highlights the potential of sequential use of MET-TKIs. Clin Cancer Res; 23(16); 4929-37. ©2017 AACR.
Collapse
Affiliation(s)
- Anna Li
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhou Zhang
- Burning Rock Biotech, Guangzhou, Guangdong Province, P.R. China
| | - Jian Su
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Lan-Ying Gou
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Yu Bai
- Asia Innovative Medicines and Early Development, AstraZeneca, Shanghai, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhenfan Yang
- Asia Innovative Medicines and Early Development, AstraZeneca, Shanghai, China
| | - Han Han-Zhang
- Burning Rock Biotech, Guangzhou, Guangdong Province, P.R. China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Shannon Chuai
- Burning Rock Biotech, Guangzhou, Guangdong Province, P.R. China
| | - Qi Zhang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhi Xie
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Hongfei Gao
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Huajun Chen
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhen Wang
- Asia Innovative Medicines and Early Development, AstraZeneca, Shanghai, China
| | - Zheng Wang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Bin-Chao Wang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Bin Gan
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhi-Hong Chen
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Ben-Yuan Jiang
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Si-Pei Wu
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Si-Yang Liu
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Chong-Rui Xu
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong General Hospital, and Guangdong Academy of Medical Sciences, Guangzhou, P.R. China.
| |
Collapse
|
77
|
Targeting c-MET in gastrointestinal tumours: rationale, opportunities and challenges. Nat Rev Clin Oncol 2017; 14:562-576. [PMID: 28374784 DOI: 10.1038/nrclinonc.2017.40] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Data from many preclinical studies, including those using cellular models of colorectal, gastric, gastro-oesophageal and gastro-oesophageal junction cancers, indicate that the hepatocyte growth factor (HGF)-hepatocyte growth factor receptor (c-MET) pathway is vital for the growth, survival and invasive potential of gastrointestinal cancers. Following the availability of data from these various studies, and data on c-MET expression as a biomarker that indicates a poor prognosis in patients with gastrointestinal cancer and increased c-MET expression, inhibitors targeting this pathway have entered the clinic in the past decade. However, the design of clinical trials that incorporate the use of HGF/c-MET inhibitors in their most appropriate genetic and molecular context remains crucial. Recognizing and responding to this challenge, the European Commission funded Framework 7 MErCuRIC programme is running a biomarker-enriched clinical trial investigating the efficacy of combined c-MET/MEK inhibition in patients with RAS-mutant or RAS-wild-type metastatic colorectal cancer with aberrant c-MET expression. The design of this trial enables the continued refinement of the predictive biomarker and co-development of companion diagnostics. In this Review, we focus on advances in our understanding of inhibition of the HGF/c-MET pathway in patients with gastro-intestinal cancers, the prominent challenges facing the clinical translation and implementation of agents targeting HGF/c-MET, and discuss the various efforts, and associated obstacles to the discovery and validation of biomarkers that will enable patient stratification in this context.
Collapse
|
78
|
Shu Y, Wu X, Tong X, Wang X, Chang Z, Mao Y, Chen X, Sun J, Wang Z, Hong Z, Zhu L, Zhu C, Chen J, Liang Y, Shao H, Shao YW. Circulating Tumor DNA Mutation Profiling by Targeted Next Generation Sequencing Provides Guidance for Personalized Treatments in Multiple Cancer Types. Sci Rep 2017; 7:583. [PMID: 28373672 PMCID: PMC5428730 DOI: 10.1038/s41598-017-00520-1] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 03/03/2017] [Indexed: 12/12/2022] Open
Abstract
Cancer is a disease of complex genetic alterations, and comprehensive genetic diagnosis is beneficial to match each patient to appropriate therapy. However, acquisition of representative tumor samples is invasive and sometimes impossible. Circulating tumor DNA (ctDNA) is a promising tool to use as a non-invasive biomarker for cancer mutation profiling. Here we implemented targeted next generation sequencing (NGS) with a customized gene panel of 382 cancer-relevant genes on 605 ctDNA samples in multiple cancer types. Overall, tumor-specific mutations were identified in 87% of ctDNA samples, with mutation spectra highly concordant with their matched tumor tissues. 71% of patients had at least one clinically-actionable mutation, 76% of which have suggested drugs approved or in clinical trials. In particular, our study reveals a unique mutation spectrum in Chinese lung cancer patients which could be used to guide treatment decisions and monitor drug-resistant mutations. Taken together, our study demonstrated the feasibility of clinically-useful targeted NGS-based ctDNA mutation profiling to guide treatment decisions in cancer.
Collapse
Affiliation(s)
- Yongqian Shu
- Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xue Wu
- Geneseeq Technology Inc., Toronto, Ontario, Canada
| | | | - Xiaonan Wang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Zhili Chang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Yu Mao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Xiaofeng Chen
- Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Sun
- Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhenxin Wang
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhuan Hong
- Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Nanjing, Jiangsu, China
| | - Liangjun Zhu
- Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Nanjing, Jiangsu, China
| | - Chunrong Zhu
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Chen
- Department of Chemoradiotherapy, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Ying Liang
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Huawu Shao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China.,Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, China
| | - Yang W Shao
- Geneseeq Technology Inc., Toronto, Ontario, Canada.
| |
Collapse
|
79
|
Farrell PJ, Matuszkiewicz J, Balakrishna D, Pandya S, Hixon MS, Kamran R, Chu S, Lawson JD, Okada K, Hori A, Mizutani A, Iwata H, de Jong R, Hibner B, Vincent P. MET Tyrosine Kinase Inhibition Enhances the Antitumor Efficacy of an HGF Antibody. Mol Cancer Ther 2017; 16:1269-1278. [PMID: 28341789 DOI: 10.1158/1535-7163.mct-16-0771] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/13/2016] [Accepted: 03/15/2017] [Indexed: 11/16/2022]
Abstract
Receptor tyrosine kinase therapies have proven to be efficacious in specific cancer patient populations; however, a significant limitation of tyrosine kinase inhibitor (TKI) treatment is the emergence of resistance mechanisms leading to a transient, partial, or complete lack of response. Combination therapies using agents with synergistic activity have potential to improve response and reduce acquired resistance. Chemoreagent or TKI treatment can lead to increased expression of hepatocyte growth factor (HGF) and/or MET, and this effect correlates with increased metastasis and poor prognosis. Despite MET's role in resistance and cancer biology, MET TKI monotherapy has yielded disappointing clinical responses. In this study, we describe the biological activity of a selective, oral MET TKI with slow off-rate and its synergistic antitumor effects when combined with an anti-HGF antibody. We evaluated the combined action of simultaneously neutralizing HGF ligand and inhibiting MET kinase activity in two cancer xenograft models that exhibit autocrine HGF/MET activation. The combination therapy results in additive antitumor activity in KP4 pancreatic tumors and synergistic activity in U-87MG glioblastoma tumors. Pharmacodynamic characterization of biomarkers that correlate with combination synergy reveal that monotherapies induce an increase in the total MET protein, whereas combination therapy significantly reduces total MET protein levels and phosphorylation of 4E-BP1. These results hold promise that dual targeting of HGF and MET by combining extracellular ligand inhibitors with intracellular MET TKIs could be an effective intervention strategy for cancer patients who have acquired resistance that is dependent on total MET protein. Mol Cancer Ther; 16(7); 1269-78. ©2017 AACR.
Collapse
Affiliation(s)
- Pamela J Farrell
- Department of Biological Sciences, Takeda California, San Diego, California.
| | | | | | - Shweta Pandya
- Department of Biological Sciences, Takeda California, San Diego, California
| | - Mark S Hixon
- Department of Biological Sciences, Takeda California, San Diego, California
| | - Ruhi Kamran
- Department of Biological Sciences, Takeda California, San Diego, California
| | - Shaosong Chu
- Department of Chemistry, Takeda California, San Diego, California
| | - J David Lawson
- Department of Computational Sciences and Crystallography, Takeda California, San Diego, California
| | - Kengo Okada
- Pharmaceutical Research Division, Takeda Pharmaceutical Companies Ltd, Shonan, Japan
| | - Akira Hori
- Pharmaceutical Research Division, Takeda Pharmaceutical Companies Ltd, Shonan, Japan
| | - Akio Mizutani
- Pharmaceutical Research Division, Takeda Pharmaceutical Companies Ltd, Shonan, Japan
| | - Hidehisa Iwata
- Pharmaceutical Research Division, Takeda Pharmaceutical Companies Ltd, Shonan, Japan
| | - Ron de Jong
- Department of Biological Sciences, Takeda California, San Diego, California
| | - Barbara Hibner
- Oncology Biology, Takeda Boston, Cambridge, Massachusetts
| | - Patrick Vincent
- Department of Biological Sciences, Takeda California, San Diego, California
| |
Collapse
|
80
|
Cortot AB, Kherrouche Z, Descarpentries C, Wislez M, Baldacci S, Furlan A, Tulasne D. Exon 14 Deleted MET Receptor as a New Biomarker and Target in Cancers. J Natl Cancer Inst 2017; 109:2982828. [DOI: 10.1093/jnci/djw262] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 10/04/2016] [Indexed: 01/07/2023] Open
|
81
|
Abstract
An increasing number of patients are diagnosed with esophageal cancer at an advanced stages, and only a small group of them can benefit from the traditional chemotherapy and radiotherapy. So far, multiple monoclonal antibodies and tyrosine kinase inhibitors have been developed, alone or in combination with traditional therapy, to improve the prognosis of patients with advanced esophageal cancer. This review summarizes the recent advances of targeted therapies against EGFR, HER2, VEGFR and c-MET in esophageal cancer. More clinical trials should be performed to evaluate the efficacy and safety of various targeted therapy regimens. Future basic research should focus on investigating the molecular mechanisms of therapeutic targets in esophageal cancer.
Collapse
Affiliation(s)
- Lei Zhang
- a Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China.,b Department of General Surgery , NO. 406 Hospital, Da Lian , China
| | - Jiaojiao Ma
- a Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Yu Han
- c Department of Otolaryngology , Xijing Hospital, Fourth Military Medical University , Xi'an , China
| | - Jinqiang Liu
- a Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Wei Zhou
- a Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Liu Hong
- a Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Daiming Fan
- a Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| |
Collapse
|
82
|
Ko B, He T, Gadgeel S, Halmos B. MET/HGF pathway activation as a paradigm of resistance to targeted therapies. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:4. [PMID: 28164089 DOI: 10.21037/atm.2016.12.09] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Resistance to targeted therapeutics is a key issue limiting the long-term utility of these medications in the management of molecularly selected subsets of cancer patients, including patients with non-small cell lung cancer harboring oncogenic alterations affecting EGFR, ALK and other genes. Bypass resistance mediated by activation of MET kinase has emerged as a frequent, validated and pivotal resistance mechanism in multiple types of cancers. Biochemical understanding is accumulating to explain the unique role of MET in such bypass pathways, providing alternate downstream activation opportunities and intricate interactions during epithelial-mesenchymal transitions. Multiple diagnostic testing platforms have become available for selecting appropriate patients for MET targeting in a variety of settings. Importantly, in light of the failures of several earlier clinical studies of MET targeting agents, a large array of recent and current MET-focused trials are incorporating stricter patient selection and more robust predictive biomarkers providing hope for validation of MET targeting as a clinically impactful strategy.
Collapse
Affiliation(s)
- Brian Ko
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Tianfang He
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Shirish Gadgeel
- Department of Oncology, Barbara Ann Karmanos Cancer Institute/Wayne State University, Detroit, MI 48201, USA
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY 10467, USA
| |
Collapse
|
83
|
Yap TA, Popat S. Targeting MET Exon 14 Skipping Alterations: Has Lung Cancer MET Its Match? J Thorac Oncol 2017; 12:12-14. [DOI: 10.1016/j.jtho.2016.10.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 10/30/2016] [Indexed: 10/20/2022]
|
84
|
Ou SHI, Young L, Schrock AB, Johnson A, Klempner SJ, Zhu VW, Miller VA, Ali SM. Emergence of Preexisting MET Y1230C Mutation as a Resistance Mechanism to Crizotinib in NSCLC with MET Exon 14 Skipping. J Thorac Oncol 2017; 12:137-140. [DOI: 10.1016/j.jtho.2016.09.119] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/13/2016] [Accepted: 09/13/2016] [Indexed: 11/15/2022]
|
85
|
Responses to Crizotinib Can Occur in High-Level MET -Amplified Non–Small Cell Lung Cancer Independent of MET Exon 14 Alterations. J Thorac Oncol 2017; 12:141-144. [DOI: 10.1016/j.jtho.2016.09.116] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/11/2016] [Accepted: 09/05/2016] [Indexed: 01/01/2023]
|
86
|
Miekus K. The Met tyrosine kinase receptor as a therapeutic target and a potential cancer stem cell factor responsible for therapy resistance (Review). Oncol Rep 2016; 37:647-656. [PMID: 27959446 DOI: 10.3892/or.2016.5297] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/17/2016] [Indexed: 11/05/2022] Open
Abstract
The MET tyrosine kinase receptor plays an important role during tumor development and progression being responsible for proliferation, morphogenetic transformation, cell motility and invasiveness. High expression of the MET receptor has been shown to correlate with increased tumor growth and metastasis, poor prognosis and resistance to radiotherapy. Moreover, MET expression and activation has been shown to be associated with therapy resistance. The occurrence of resistance to targeted therapy might be related to the presence of cancer stem cells (CSCs). CSCs are a subpopulation of cells in the tumor that possess the ability of self-renewal, clonogenicity, radioresistance and self-sustained protection from apoptosis. Recently, MET has been postulated as an essential factor supporting the functional stem cell phenotype in some tumors and as a CSC factor is believed to be responsible for therapy resistance. This review presents the results from recent studies identifying MET as a potential marker of CSCs and tumor initiating cells, demonstrating pivotal role of MET in supporting stem cell phenotype and indicating the role of MET in acquiring resistance to antitumor therapy.
Collapse
Affiliation(s)
- Katarzyna Miekus
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Cracow, Poland
| |
Collapse
|
87
|
Reungwetwattana T, Liang Y, Zhu V, Ou SHI. The race to target MET exon 14 skipping alterations in non-small cell lung cancer: The Why, the How, the Who, the Unknown, and the Inevitable. Lung Cancer 2016; 103:27-37. [PMID: 28024693 DOI: 10.1016/j.lungcan.2016.11.011] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/11/2016] [Accepted: 11/13/2016] [Indexed: 01/29/2023]
Abstract
A number of small molecule tyrosine kinase inhibitors (TKIs) have now been approved for the treatment of non-small cell lung cancers (NSCLC), including those targeted against epidermal growth factor receptor, anaplastic lymphoma kinase, and ROS1. Despite a wealth of agents developed to target the receptor tyrosine kinase, MET, clinical outcomes have as yet been disappointing, leading to pessimism about the role of MET in the pathogenesis of NSCLC. However, in recent years, there has been a renewed interest in MET exon 14 alterations as potential drivers of lung cancer. MET exon 14 alterations, which result in increased MET protein levels due to disrupted ubiquitin-mediated degradation, occur at a prevalence of around 3% in adenocarcinomas and around 2% in other lung neoplasms, making them attractive targets for the treatment of lung cancer. At least five MET-targeted TKIs, including crizotinib, cabozantinib, capmatinib, tepotinib, and glesatinib, are being investigated clinically for patients with MET exon 14 altered-NSCLC. A further two compounds have shown activity in preclinical models. In this article, we review the current clinical and preclinical data available for these TKIs, along with a number of other potential therapeutic options, including antibodies and immunotherapy. A number of questions remain unanswered regarding the future of MET TKIs, but unfortunately, the development of resistance to targeted therapies is inevitable. Resistance is expected to arise as a result of receptor tyrosine kinase mutation or from upregulation of MET ligand expression; potential strategies to overcome resistance are proposed.
Collapse
Affiliation(s)
- Thanyanan Reungwetwattana
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Ying Liang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Viola Zhu
- Long Beach Veterans Administration Hospital, Long Beach, CA 90822, USA; Chao Family Comprehensive Cancer Center, Department of Medicine, Division of Hematology-Oncology, University of California Irvine School of Medicine, Orange County, CA 92868, USA
| | - Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, Department of Medicine, Division of Hematology-Oncology, University of California Irvine School of Medicine, Orange County, CA 92868, USA.
| |
Collapse
|
88
|
Response and acquired resistance to crizotinib in Chinese patients with lung adenocarcinomas harboring MET Exon 14 splicing alternations. Lung Cancer 2016; 102:118-121. [PMID: 27987579 DOI: 10.1016/j.lungcan.2016.11.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/17/2016] [Accepted: 11/07/2016] [Indexed: 11/22/2022]
Abstract
Approximately 10% of lung adenocarcinomas harbor aberrations that are targetable using the approved multitargeted TKI crizotinib. MET exon 14 skipping mutation predicts for response to crizotinib in human lung adenocarcinomas. However, a substantial part of patients still has no sufficient tissue to perform genomic analysis. As a promising noninvasive biomarker and potential surrogate for the entire tumor genome, circulating tumor DNA (ctDNA) has been applied to the detection of driver gene mutations. Here we described the MET exon 14 splicing mutations in cell-free circulating-tumor DNA by next-generation sequencing (NGS) technology. Patient firstly responded to crizotinib therapy within four months, however, three acquired mutation in the MET kinase domain, D1228N/H and Y1230H, were found at the time of disease progression. To our knowledge, this is the first clinical report of three mutations simultaneously arising in a patient with MET exon 14 splicing mutation.
Collapse
|
89
|
Zhao Z, Verma V, Zhang M. Anaplastic lymphoma kinase: Role in cancer and therapy perspective. Cancer Biol Ther 2016; 16:1691-701. [PMID: 26529396 DOI: 10.1080/15384047.2015.1095407] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK) is correlated with oncogenesis in different types of cancers, such as anaplastic large cell lymphoma, lung cancer, neuroblastoma, and even breast cancer, by abnormal fusion of ALK or non-fusion ALK activation. ALK is a receptor tyrosine kinase, with a single transmembrane domain, that plays an important role in development. Upon ligand binding to the extracellular domain, the receptor undergoes dimerization and subsequent autophosphorylation of the intracellular kinase domain. In recent years, ALK inhibitors have been developed for cancer treatment. These inhibitors target ALK activity and show effectiveness in ALK-positive non-small cell lung cancer. However, acquired treatment resistance makes the future of this therapy unclear; new strategies are underway to overcome the limitations of current ALK inhibitors.
Collapse
Affiliation(s)
- Zhihong Zhao
- a Munroe-Meyer Institute; University of Nebraska Medical Center ; Omaha , NE , USA
| | - Vivek Verma
- b Department of Radiation Oncology ; University of Nebraska Medical Center ; Omaha , NE , USA
| | - Mutian Zhang
- b Department of Radiation Oncology ; University of Nebraska Medical Center ; Omaha , NE , USA
| |
Collapse
|
90
|
Yamaoka T, Ohmori T, Ohba M, Arata S, Kishino Y, Murata Y, Kusumoto S, Ishida H, Shirai T, Hirose T, Ohnishi T, Sasaki Y. Acquired Resistance Mechanisms to Combination Met-TKI/EGFR-TKI Exposure in Met-Amplified EGFR-TKI-Resistant Lung Adenocarcinoma Harboring an Activating EGFR Mutation. Mol Cancer Ther 2016; 15:3040-3054. [PMID: 27612490 DOI: 10.1158/1535-7163.mct-16-0313] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/03/2016] [Accepted: 08/06/2016] [Indexed: 11/16/2022]
Abstract
Met-amplified EGFR-tyrosine kinase inhibitor (TKI)-resistant non-small cell lung cancer (NSCLC) harboring an activating EGFR mutation is responsive to concurrent EGFR-TKI and Met-TKI treatment in a preclinical model. Here, we determined that Met-amplified gefitinib-resistant cells acquire dual resistance to inhibition of EGFR and Met tyrosine kinase activities. PC-9 lung adenocarcinoma cells harboring 15-bp deletions (Del E746_A750) in EGFR exon 19 were treated with increasing concentrations of the Met-TKI PHA665752 and 1 μmol/L gefitinib for 1 year; three resistant clones were established via Met amplification. The three dual-resistance cell lines (PC-9DR2, PC-9DR4, and PC-9DR6, designated as DR2, DR4, and DR6, respectively) exhibited different mechanisms for evading both EGFR and Met inhibition. None of the clones harbored a secondary mutation of EGFR T790M or a Met mutation. Insulin-like growth factor (IGF)/IGF1 receptor activation in DR2 and DR4 cells acted as a bypass signaling pathway. Met expression was attenuated to a greater extent in DR2 than in PC-9 cells, but was maintained in DR4 cells by overexpression of IGF-binding protein 3. In DR6 cells, Met was further amplified by association with HSP90, which protected Met from degradation and induced SET and MYND domain-containing 3 (SMYD3)-mediated Met transcription. This is the first report describing the acquisition of dual resistance mechanisms in NSCLC harboring an activating EGFR mutation to Met-TKI and EGFR-TKI following previous EGFR-TKI treatment. These results might inform the development of more effective therapeutic strategies for NSCLC treatment. Mol Cancer Ther; 15(12); 3040-54. ©2016 AACR.
Collapse
Affiliation(s)
| | - Tohru Ohmori
- Institute of Molecular Oncology, Showa University, Tokyo, Japan
| | - Motoi Ohba
- Institute of Molecular Oncology, Showa University, Tokyo, Japan
| | - Satoru Arata
- Institute of Molecular Oncology, Showa University, Tokyo, Japan.,Center for Biotechnology, Showa University, Tokyo, Japan
| | - Yasunari Kishino
- Division of Allergology and Respiratory Medicine, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yasunori Murata
- Division of Allergology and Respiratory Medicine, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Sojiro Kusumoto
- Division of Allergology and Respiratory Medicine, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hiroo Ishida
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takao Shirai
- Division of Allergology and Respiratory Medicine, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takashi Hirose
- Division of Allergology and Respiratory Medicine, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Tsukasa Ohnishi
- Division of Allergology and Respiratory Medicine, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yasutsuna Sasaki
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
91
|
Design, synthesis and biological evaluation of c-Met kinase inhibitors bearing 2-oxo-1,2-dihydroquinoline scaffold. Bioorg Med Chem Lett 2016; 26:4483-4486. [DOI: 10.1016/j.bmcl.2016.07.077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 07/12/2016] [Accepted: 07/29/2016] [Indexed: 01/03/2023]
|
92
|
Dual MET/EGFR therapy leads to complete response and resistance prevention in a MET-amplified gastroesophageal xenopatient cohort. Oncogene 2016; 36:1200-1210. [PMID: 27524418 DOI: 10.1038/onc.2016.283] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/10/2016] [Accepted: 06/23/2016] [Indexed: 12/12/2022]
Abstract
Amplification of the MET oncogene occurs in 2-4% of gastroesophageal cancers and defines a small and aggressive subset of tumors. Although in vitro studies have given very promising results, clinical trials with MET inhibitors have been disappointing, showing few and short lasting responses. The aim of the work was to exploit a MET-amplified patient-derived xenograft model to optimize anti-MET therapeutic strategies in gastroesophageal cancer. We found that despite the high MET amplification level (26 gene copies), in the absence of qualitative or quantitative alterations of EGFR, MET inhibitors induced only tumor growth inhibition, whereas dual MET/EGFR inhibition led to complete tumor regression. Importantly, the combo treatment completely prevented the onset of resistance, which quite rapidly appeared in tumors treated with MET monotherapy. We found that this secondary resistance was due to EGFR activation and could be overcome by dual MET/EGFR inhibition. Similar results were also obtained in a MET-addicted, established gastric cancer cell line. In vitro experiments performed on tumor-derived primary cells confirmed that MET inhibitors were not able to abrogate the activation of downstream transducers and that only the combined MET/EGFR treatment completely shut off the signaling. Previously reported cases, as well as those described here, showed only partial and transient sensitivity to anti-MET therapy. The finding that combined anti-MET/EGFR therapy-even in the absence of EGFR genetic alterations-induced complete and durable response, represents a proof of concept and guarantees further investigations, opening a new perspective of treatment for these patients.
Collapse
|
93
|
Acquired Resistance to Crizotinib in NSCLC with MET Exon 14 Skipping. J Thorac Oncol 2016; 11:1242-1245. [PMID: 27343442 DOI: 10.1016/j.jtho.2016.06.013] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 06/17/2016] [Accepted: 06/17/2016] [Indexed: 11/24/2022]
Abstract
INTRODUCTION MET proto-oncogene, receptor tyrosine kinase gene (MET) exon 14 skipping is a targetable alteration in lung cancer. Treatment with MET proto-oncogene, receptor tyrosine kinase inhibitor can cause dramatic responses in patients whose cancers have MET exon 14 skipping. Little is known, however, about acquired resistance in patients with MET exon 14 skipping. METHODS Biopsy specimens obtained at baseline and at the time of progression for a patient being treated with crizotinib were compared using targeted next-generation sequencing to assess for mechanisms of resistance. RESULTS An acquired mutation in the MET kinase domain, D1228N, was found at time of progression on crizotinib in a patient with MET exon 14 skipping. CONCLUSIONS One potential mechanism of acquired resistance to crizotinib in patients with MET exon 14 skipping is through second-site mutations in the MET gene. Understanding mechanisms of resistance will be important in optimizing therapy in these patients.
Collapse
|
94
|
Salvi S, Varesano S, Boccardo S, Ravetti JL, Canessa PA, Pistillo MP, Ferro P, Fedeli F, Roncella S. EGFR Status in Mesothelioma: Possible Implications for the Efficacy of Anti-EGFR and Anti-MET Therapies. J Thorac Oncol 2016; 11:e78-80. [DOI: 10.1016/j.jtho.2016.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 03/07/2016] [Indexed: 10/21/2022]
|
95
|
An N, Xiong Y, LaRue AC, Kraft AS, Cen B. Activation of Pim Kinases Is Sufficient to Promote Resistance to MET Small-Molecule Inhibitors. Cancer Res 2016; 75:5318-28. [PMID: 26670562 DOI: 10.1158/0008-5472.can-15-0544] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesenchymal-epithelial transition (MET) blockade offers a new targeted therapy particularly in those cancers with MET amplification. However, the efficacy and the duration of the response to MET inhibitors are limited by the emergence of drug resistance. Here, we report that resistance to small-molecule inhibitors of MET can arise from increased expression of the prosurvival Pim protein kinases. This resistance mechanism was documented in non-small cell lung cancer and gastric cancer cells with MET amplification. Inhibition of Pim kinases enhanced cell death triggered by short-term treatment with MET inhibitors. Pim kinases control the translation of antiapoptotic protein Bcl-2 at an internal ribosome entry site and this mechanism was identified as the basis for Pim-mediated resistance to MET inhibitors. Protein synthesis was increased in drug-resistant cells, secondary to a Pim-mediated increase in cap-independent translation. In cells rendered drug resistant by chronic treatment with MET inhibitors, genetic or pharmacologic inhibition of Pim kinases was sufficient to restore sensitivity in vitro and in vivo. Taken together, our results rationalize Pim inhibition as a strategy to augment responses and blunt acquired resistance to MET inhibitors in cancer.
Collapse
Affiliation(s)
- Ningfei An
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina. The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Ying Xiong
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Amanda C LaRue
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina. Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | | | - Bo Cen
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina. The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
96
|
Ohashi Y, Okamura M, Hirosawa A, Tamaki N, Akatsuka A, Wu KM, Choi HW, Yoshimatsu K, Shiina I, Yamori T, Dan S. M-COPA, a Golgi Disruptor, Inhibits Cell Surface Expression of MET Protein and Exhibits Antitumor Activity against MET-Addicted Gastric Cancers. Cancer Res 2016; 76:3895-903. [PMID: 27197184 DOI: 10.1158/0008-5472.can-15-2220] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 03/28/2016] [Indexed: 12/31/2022]
Abstract
The Golgi apparatus is responsible for transporting, processing, and sorting numerous proteins in the cell, including cell surface-expressed receptor tyrosine kinases (RTK). The small-molecule compound M-COPA [2-methylcoprophilinamide (AMF-26)] disrupts the Golgi apparatus by inhibiting the activation of Arf1, resulting in suppression of tumor growth. Here, we report an evaluation of M-COPA activity against RTK-addicted cancers, focusing specifically on human gastric cancer (GC) cells with or without MET amplification. As expected, the MET-addicted cell line MKN45 exhibited a better response to M-COPA than cell lines without MET amplification. Upon M-COPA treatment, cell surface expression of MET was downregulated with a concurrent accumulation of its precursor form. M-COPA also reduced levels of the phosphorylated form of MET along with the downstream signaling molecules Akt and S6. Similar results were obtained in additional GC cell lines with amplification of MET or the FGF receptor FGFR2 MKN45 murine xenograft experiments demonstrated the antitumor activity of M-COPA in vivo Taken together, our results offer an initial preclinical proof of concept for the use of M-COPA as a candidate treatment option for MET-addicted GC, with broader implications for targeting the Golgi apparatus as a novel cancer therapeutic approach. Cancer Res; 76(13); 3895-903. ©2016 AACR.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Apoptosis/drug effects
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Case-Control Studies
- Cell Proliferation/drug effects
- Female
- Follow-Up Studies
- Golgi Apparatus/drug effects
- Golgi Apparatus/metabolism
- Golgi Apparatus/pathology
- Humans
- Immunoenzyme Techniques
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Naphthols/pharmacology
- Neoplasm Staging
- Phosphorylation/drug effects
- Prognosis
- Proto-Oncogene Proteins c-met/genetics
- Proto-Oncogene Proteins c-met/metabolism
- Pyridines/pharmacology
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Signal Transduction/drug effects
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yoshimi Ohashi
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mutsumi Okamura
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Asaka Hirosawa
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Naomi Tamaki
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akinobu Akatsuka
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kuo-Ming Wu
- Next Generation Systems, Eisai Inc., Andover, Massachusetts
| | | | | | - Isamu Shiina
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Tokyo, Japan
| | - Takao Yamori
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
97
|
Cui J, Xia T, Xie D, Gao Y, Jia Z, Wei D, Wang L, Huang S, Quan M, Xie K. HGF/Met and FOXM1 form a positive feedback loop and render pancreatic cancer cells resistance to Met inhibition and aggressive phenotypes. Oncogene 2016; 35:4708-18. [PMID: 26876216 PMCID: PMC4985506 DOI: 10.1038/onc.2016.14] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 11/30/2015] [Accepted: 12/27/2015] [Indexed: 02/06/2023]
Abstract
Purpose Hepatocyte growth factor (HGF)/Met signaling plays critical roles in pancreatic ductal adenocarcinoma (PDA) development and progression and is considered a potential therapeutic target for this disease. However, the mechanism of aberrant activation of HGF/Met signaling and resistance to Met inhibition in PDA remains unclear. Experimental Design The mechanistic role of cross-talk between Forkhead box M1 (FOXM1) and HGF/Met signaling in promotion of PDA growth and resistance to Met inhibition was examined using cell culture, molecular biology and mouse models; and the relevance of our experimental and mechanistic findings were validated using human PDA tissues. Results Met was markedly overexpressed in both PDA cell lines and pancreatic tumor specimens, and the expression of Met correlated directly with that of FOXM1 in human tumor specimens. Mechanistically, FOXM1 bound to the promoter region of the Met gene and transcriptionally increased the expression of Met. Increased expression of FOXM1 enhanced the activation of HGF/Met signaling and its downstream pathways, including RAS/extracellular signal-regulated kinase 1/2, phosphoinositide 3-kinase/AKT, and signal transducer and activator of transcription 3. Furthermore, activation of HGF/Met signaling increased the expression and transcriptional activity of FOXM1, and the cross-talk between FOXM1 and HGF/Met signaling promoted PDA growth and resistance to Met inhibition. Conclusions Collectively, our findings identified a positive feedback loop formed by FOXM1 and HGF/Met and revealed that this loop is a potentially effective therapeutic target for PDA.
Collapse
Affiliation(s)
- J Cui
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - T Xia
- Department of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - D Xie
- Department of Oncology, Shanghai Tongji University Affiliated East Hospital, Shanghai, People's Republic of China
| | - Y Gao
- Department of Oncology, Shanghai Tongji University Affiliated East Hospital, Shanghai, People's Republic of China
| | - Z Jia
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - D Wei
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Wang
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - S Huang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Quan
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Oncology, Shanghai Tongji University Affiliated East Hospital, Shanghai, People's Republic of China
| | - K Xie
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
98
|
Lee J, Tran P, Klempner SJ. Targeting the MET Pathway in Gastric and Oesophageal Cancers: Refining the Optimal Approach. Clin Oncol (R Coll Radiol) 2016; 28:e35-44. [PMID: 26880063 DOI: 10.1016/j.clon.2016.01.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/09/2015] [Accepted: 12/10/2015] [Indexed: 12/21/2022]
Abstract
Gastric and oesophageal cancers are a major cause of global cancer-related morbidity and mortality. Improvements in treatment for locoregional and metastatic gastric and oesophageal cancer have been incremental and the overall prognosis remains poor. Increasingly, molecular classification has identified recurrent, therapeutically relevant, somatic alterations in gastroesophageal malignancies. However, other than ERBB2 amplification, molecularly directed therapies have not translated to improved survival. Amplification of the receptor tyrosine kinase MET is found in about 5% of gastroesophageal cancers and represents an oncogenic driver and therapeutic target. Small series have shown activity of MET-directed tyrosine kinase inhibitors, but the clinical benefit of anti-MET antibodies has been disappointing. Here we discuss the MET pathway in gastroesophageal cancers, the clinical data for MET small molecule tyrosine kinase inhibitors, anti-MET antibodies and future clinical directions for targeting MET in gastric and oesophageal cancers. To our knowledge, this is the most comprehensive review of the clinical experience with MET-directed therapies in gastric and oesophageal cancers.
Collapse
Affiliation(s)
- J Lee
- Department of Medicine, University of California Irvine, Orange, CA, USA
| | - P Tran
- Division of Hematology-Oncology, University of California Irvine, Orange, CA, USA
| | - S J Klempner
- Division of Hematology-Oncology, University of California Irvine, Orange, CA, USA.
| |
Collapse
|
99
|
Brady SW, Zhang J, Tsai MH, Yu D. PI3K-independent mTOR activation promotes lapatinib resistance and IAP expression that can be effectively reversed by mTOR and Hsp90 inhibition. Cancer Biol Ther 2016; 16:402-11. [PMID: 25692408 DOI: 10.1080/15384047.2014.1002693] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Although HER2 targeted therapies have substantially improved outcomes in HER2 overexpressing (HER2+) breast cancer, resistance to these therapies remains a clinical challenge. To better understand the mechanisms of resistance to lapatinib, a HER2 and EGFR dual kinase inhibitor, we treated HER2+ breast cancer cells with lapatinib for an extended period to generate a lapatinib-resistant (LapR) cell line model and examined cancer-promoting signaling activation in LapR cells. We found that LapR cells possess enhanced mTOR activation, which was independent of PI3K and other known mTOR activators. Lapatinib resistance could be reversed by mTOR kinase inhibition. Intriguingly, LapR cells had constitutive cytosolic cytochrome C, indicating that LapR cells suppress lapatinib-induced apoptosis downstream of cytochrome C release from mitochondria into the cytosol rather than by preventing its release into the cytosol. Consistent with this notion, LapR cells possessed increased levels of 2 of the inhibitors of apoptosis (IAPs), survivin and c-IAP-2, which are reported to block caspase activation downstream of cytosolic cytochrome C release. Further, treatment with the mTOR kinase inhibitor AZD8055 or the Hsp90 inhibitor 17-AAG reversed expression of IAPs and overcame lapatinib resistance in LapR cells. Together, these data suggest that suppression of apoptosis downstream of cytosolic cytochrome C release, possibly through increased expression of IAPs or other caspase-suppressing proteins, may promote lapatinib resistance. Further, PI3K is thought to be the main driver of lapatinib resistance, but our findings indicate that PI3K inhibitors may be ineffective in some lapatinib-resistant HER2+ breast cancers with PI3K-independent activation of mTOR kinase, which may instead benefit from mTOR or Hsp90 inhibitors.
Collapse
Affiliation(s)
- Samuel W Brady
- a Department of Molecular and Cellular Oncology ; The University of Texas MD Anderson Cancer Center ; Houston , TX USA
| | | | | | | |
Collapse
|
100
|
Kim DC, Park KR, Jeong YJ, Yoon H, Ahn MJ, Rho GJ, Lee J, Gong YD, Han SY. Resistance to the c-Met inhibitor KRC-108 induces the epithelial transition of gastric cancer cells. Oncol Lett 2015; 11:991-997. [PMID: 26893681 PMCID: PMC4734112 DOI: 10.3892/ol.2015.4029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 08/17/2015] [Indexed: 12/16/2022] Open
Abstract
Investigation of the mechanisms of resistance to targeted therapies is essential as resistance acquired during treatment may lead to relapse or refractoriness to the therapy. Our previous study identified the small molecule KRC-108 as a result of efforts to find an anticancer agent with c-Met-inhibitory activity. In the present study, the changes accompanying resistance to KRC-108 were investigated in the gastric cancer cell line MKN-45 and its KRC-108-resistant clones by western blot and immunofluorescence analyses. Increased expression of the c-Met protein was observed in KRC-108-resistant cells compared with that of the parental cells, and the phosphorylation of c-Met also increased in cell lines resistant to KRC-108. Resistance to the c-Met inhibitor was associated with cell morphological changes: MKN-45 parental cells, which had a round and poorly differentiated morphology, were altered to exhibit an epithelial cell-like phenotype in KRC-108-resistant clones. Consistent with the transition to an epithelial morphology, the expression of E-cadherin was increased in resistant cells. Using immunoprecipitation, an interaction between E-cadherin and the c-Met protein was observed in the KRC-108-resistant cells. Immunohistochemical analysis of human gastric carcinoma tissues revealed the co-expression of E-cadherin and c-Met. These results suggest that the epithelial transition in KRC-108-resistant cells is mediated by recruiting E-cadherin to c-Met protein. Thus, the present study identified a mechanism used by cancer cells to confer resistance to anticancer agents.
Collapse
Affiliation(s)
- Dong Chul Kim
- Department of Pathology, School of Medicine, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea
| | - Kyeong Ryang Park
- College of Pharmacy, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea; Research Institute of Life Sciences, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea
| | - Yeon Ji Jeong
- College of Pharmacy, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea; Research Institute of Life Sciences, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea
| | - Hyonok Yoon
- College of Pharmacy, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea; Research Institute of Life Sciences, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea
| | - Mi-Jeong Ahn
- College of Pharmacy, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea; Research Institute of Life Sciences, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea
| | - Gyu-Jin Rho
- Department of Theriogenology and Biotechnology, School of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea
| | - Jongkook Lee
- College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 200-701, Republic of Korea
| | - Young-Dae Gong
- Innovative Drug Library Research Center, Dongguk University, Seoul 100-715, Republic of Korea
| | - Sun-Young Han
- College of Pharmacy, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea; Research Institute of Life Sciences, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea
| |
Collapse
|