101
|
Choi J, Lee HE, Kim MA, Jang BG, Lee HS, Kim WH. Analysis of MET mRNA expression in gastric cancers using RNA in situ hybridization assay: its clinical implication and comparison with immunohistochemistry and silver in situ hybridization. PLoS One 2014; 9:e111658. [PMID: 25364819 PMCID: PMC4218795 DOI: 10.1371/journal.pone.0111658] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 10/06/2014] [Indexed: 02/07/2023] Open
Abstract
We investigated MET mRNA expression status using RNA in situ hybridization (ISH) technique in primary and metastatic lesions of 535 surgically resected gastric carcinoma (GC) cases. We compared the results with those of immunohistochemistry and silver in situ hybridization, and examined the association with clinicopathologic characteristics and prognosis. Among 535 primary GCs, 391 (73.1%) were scored 0, 87 (16.3%) were scored 1, 38 (7.1%) were scored 2, 12 (2.2%) were scored 3 and 7 (1.3%) were scored 4 by RNA ISH. High MET mRNA expression (score ≥3) was associated with lymph node metastasis (P = .014), distant metastasis (P = .001), and higher TNM stage (P<.001). MET mRNA expression was correlated with protein expression (r = 0.398; P<.001) and gene copy number (r = 0.345; P<.001). The patients showing high-MET mRNA in primary or metastatic lesions had shorter overall survival than those showing low-MET mRNA (primary tumors, P = .002; metastatic lymph nodes, P<.001). The patients showing positive conversion of MET mRNA status in metastatic lymph node had shorter overall survival than those with no conversion (P = .011). Multivariate analysis demonstrated that high MET mRNA expression in metastatic lymph node was an independent prognostic factor for overall survival (P = .007). Therefore, this study suggests that MET mRNA expression assessed by RNA ISH could be useful as a potential marker to identify MET oncogene-addicted GC.
Collapse
Affiliation(s)
- Jiwoon Choi
- Department of Pathology, Seoul National University College of Medicine, Jongno-gu, Seoul, Korea
| | - Hee Eun Lee
- Department of Pathology, Seoul National University College of Medicine, Jongno-gu, Seoul, Korea
| | - Min A. Kim
- Department of Pathology, Seoul National University College of Medicine, Jongno-gu, Seoul, Korea
| | - Bo Gun Jang
- Department of Pathology, Seoul National University College of Medicine, Jongno-gu, Seoul, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Gyeonggi, Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Jongno-gu, Seoul, Korea
- * E-mail:
| |
Collapse
|
102
|
Abdel-Rahman O. Targeting the hepatocyte growth factor/mesenchymal epithelial transition pathway in gastric cancer: biological rationale and clinical applications. Expert Rev Anticancer Ther 2014; 15:235-45. [PMID: 25353620 DOI: 10.1586/14737140.2014.974564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Advanced gastric cancer (GC) is a dreadful disease with a poor prognosis and the majority of patients die within 1 year of diagnosis. In the past decade, important signaling pathways promoting tumor proliferation and aggressiveness have been evaluated; the hepatocyte growth factor/mesenchymal epithelial transition (MET) pathway is one of the most promising pathways in that regard. This pathway has been evaluated in preclinical and early clinical settings of GC. From the very early studies, MET expression has been recognized as an important poor prognostic marker in GC. However, only after the development of MET-targeting agents, it became important in terms of antitumor therapy with the clinical evaluation of several MET-targeting agents in GC. The results of the ongoing multicenter studies evaluating MET-targeting agents are eagerly awaited as they may improve our understanding of the precise role of these agents in the treatment armamentarium of advanced GC.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Department of Clinical Oncology, Faculty of Medicine, Ain Shams University, Cairo, 113331, Egypt
| |
Collapse
|
103
|
Vita FD, Martino ND, Fabozzi A, Laterza MM, Ventriglia J, Savastano B, Petrillo A, Gambardella V, Sforza V, Marano L, Auricchio A, Galizia G, Ciardiello F, Orditura M. Clinical management of advanced gastric cancer: the role of new molecular drugs. World J Gastroenterol 2014; 20:14537-58. [PMID: 25356019 PMCID: PMC4209522 DOI: 10.3748/wjg.v20.i40.14537] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/19/2014] [Accepted: 06/02/2014] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is the fourth most common malignant neoplasm and the second leading cause of death for cancer in Western countries with more than 20000 new cases yearly diagnosed in the United States. Surgery represents the main approach for this disease but, notwithstanding the advances in surgical techniques, we observed a minimal improvement in terms of overall survival with a significant increasing of relapsing disease rates. Despite the development of new drugs has significantly improved the effectiveness of chemotherapy, the prognosis of patients with unresectable or metastatic gastric adenocarcinoma remains poor. Recently, several molecular target agents have been investigated; in particular, trastuzumab represents the first target molecule showing improvements in overall survival in human epithelial growth factor 2-positive gastric cancer patients. New molecules targeting vascular epithelial growth factor, mammalian target of rapamycin, and anti hepatocyte growth factor-c-Met pathway are also under investigation, with interesting results. Anyway, it seems necessary to select more accurately the population to treat with new agents by the identification of new biomarkers in order to optimize the results. In this paper we review the actual "scenario" of targeted treatments, also focusing on the new agents in development for gastric cancer and gastro-esophageal carcinoma, discussing their efficacy and potential applications in clinical practice.
Collapse
|
104
|
Lee D, Kim YC, Lee KM, Yoon JK, Kim YB. MET-Amplified Intramucosal Gastric Cancer Widely Metastatic after Complete Endoscopic Submucosal Dissection. Cancer Res Treat 2014; 47:120-5. [PMID: 25152190 PMCID: PMC4296859 DOI: 10.4143/crt.2013.137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 10/15/2013] [Indexed: 01/17/2023] Open
Abstract
Intramucosal gastric cancer (IGC) is associated with a very low risk of lymph node metastasis; thus it is the main candidate for minimally invasive surgical procedures, such as endoscopic submucosal dissection (ESD). Herein, we document an extraordinary case of IGC, which showed a very aggressive clinical course. A 66-year-old female underwent ESD for early gastric cancer. Histologically, the tumor consisted mainly of moderately differentiated adenocarcinoma measuring 1.6 cm in diameter, and the tumor was confined to the mucosa. Despite annual esophagogastroduodenoscopic follow-up, the tumor recurred, with wide metastasis to multiple lymph nodes and bones throughout the body after three years. Fluorescence in situ hybridization study demonstrated MET gene amplification as well as low grade polysomy 7 in both original and recurrent tumors. The clinical characteristics of metastatic IGCs and the implication of MET amplification are discussed.
Collapse
Affiliation(s)
- Dakeun Lee
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Young Chul Kim
- Department of Radiology, Ajou University School of Medicine, Suwon, Korea
| | - Kee Myung Lee
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| | - Joon-Kee Yoon
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, Korea
| | - Young-Bae Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
105
|
Kawakami H, Okamoto I, Okamoto W, Tanizaki J, Nakagawa K, Nishio K. Targeting MET Amplification as a New Oncogenic Driver. Cancers (Basel) 2014; 6:1540-52. [PMID: 25055117 PMCID: PMC4190554 DOI: 10.3390/cancers6031540] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/13/2014] [Accepted: 07/15/2014] [Indexed: 02/08/2023] Open
Abstract
Certain genetically defined cancers are dependent on a single overactive oncogene for their proliferation and survival, a phenomenon known as "oncogene addiction". A new generation of drugs that selectively target such "driver oncogenes" manifests a clinical efficacy greater than that of conventional chemotherapy in appropriate genetically defined patients. MET is a proto-oncogene that encodes a receptor tyrosine kinase, and aberrant activation of MET signaling occurs in a subset of advanced cancers as result of various genetic alterations including gene amplification, polysomy, and gene mutation. Our preclinical studies have shown that inhibition of MET signaling either with the small-molecule MET inhibitor crizotinib or by RNA interference targeted to MET mRNA resulted in marked antitumor effects in cancer cell lines with MET amplification both in vitro and in vivo. Furthermore, patients with non-small cell lung cancer or gastric cancer positive for MET amplification have shown a pronounced clinical response to crizotinib. Accumulating preclinical and clinical evidence thus suggests that MET amplification is an "oncogenic driver" and therefore a valid target for treatment. However, the prevalence of MET amplification has not been fully determined, possibly in part because of the difficulty in evaluating gene amplification. In this review, we provide a rationale for targeting this genetic alteration in cancer therapy.
Collapse
Affiliation(s)
- Hisato Kawakami
- Department of Medical Oncology, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan.
| | - Isamu Okamoto
- Department of Medical Oncology, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan.
| | - Wataru Okamoto
- Department of Medical Oncology, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan.
| | - Junko Tanizaki
- Department of Medical Oncology, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan.
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan.
| | - Kazuto Nishio
- Department of Genome Biology, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan.
| |
Collapse
|
106
|
Catenacci DVT, Liao WL, Thyparambil S, Henderson L, Xu P, Zhao L, Rambo B, Hart J, Xiao SY, Bengali K, Uzzell J, Darfler M, Krizman DB, Cecchi F, Bottaro DP, Karrison T, Veenstra TD, Hembrough T, Burrows J. Absolute quantitation of Met using mass spectrometry for clinical application: assay precision, stability, and correlation with MET gene amplification in FFPE tumor tissue. PLoS One 2014; 9:e100586. [PMID: 24983965 PMCID: PMC4077664 DOI: 10.1371/journal.pone.0100586] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 05/25/2014] [Indexed: 12/16/2022] Open
Abstract
Background Overexpression of Met tyrosine kinase receptor is associated with poor prognosis. Overexpression, and particularly MET amplification, are predictive of response to Met-specific therapy in preclinical models. Immunohistochemistry (IHC) of formalin-fixed paraffin-embedded (FFPE) tissues is currently used to select for ‘high Met’ expressing tumors for Met inhibitor trials. IHC suffers from antibody non-specificity, lack of quantitative resolution, and, when quantifying multiple proteins, inefficient use of scarce tissue. Methods After describing the development of the Liquid-Tissue-Selected Reaction Monitoring-mass spectrometry (LT-SRM-MS) Met assay, we evaluated the expression level of Met in 130 FFPE gastroesophageal cancer (GEC) tissues. We assessed the correlation of SRM Met expression to IHC and mean MET gene copy number (GCN)/nucleus or MET/CEP7 ratio by fluorescence in situ hybridization (FISH). Results Proteomic mapping of recombinant Met identified 418TEFTTALQR426 as the optimal SRM peptide. Limits of detection (LOD) and quantitation (LOQ) for this peptide were 150 and 200 amol/µg tumor protein, respectively. The assay demonstrated excellent precision and temporal stability of measurements in serial sections analyzed one year apart. Expression levels of 130 GEC tissues ranged (<150 amol/µg to 4669.5 amol/µg. High correlation was observed between SRM Met expression and both MET GCN and MET/CEP7 ratio as determined by FISH (n = 30; R2 = 0.898). IHC did not correlate well with SRM (n = 44; R2 = 0.537) nor FISH GCN (n = 31; R2 = 0.509). A Met SRM level of ≥1500 amol/µg was 100% sensitive (95% CI 0.69–1) and 100% specific (95% CI 0.92–1) for MET amplification. Conclusions The Met SRM assay measured the absolute Met levels in clinical tissues with high precision. Compared to IHC, SRM provided a quantitative and linear measurement of Met expression, reliably distinguishing between non-amplified and amplified MET tumors. These results demonstrate a novel clinical tool for efficient tumor expression profiling, potentially leading to better informed therapeutic decisions for patients with GEC.
Collapse
Affiliation(s)
- Daniel V. T. Catenacci
- Department of Medicine, Section of Hematology & Oncology, University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| | - Wei-Li Liao
- OncoPlex Diagnostics Inc., Rockville, Maryland, United States of America
| | - Sheeno Thyparambil
- OncoPlex Diagnostics Inc., Rockville, Maryland, United States of America
| | - Les Henderson
- Department of Medicine, Section of Hematology & Oncology, University of Chicago, Chicago, Illinois, United States of America
| | - Peng Xu
- Department of Medicine, Section of Hematology & Oncology, University of Chicago, Chicago, Illinois, United States of America
| | - Lei Zhao
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
| | - Brittany Rambo
- Department of Medicine, Section of Hematology & Oncology, University of Chicago, Chicago, Illinois, United States of America
| | - John Hart
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
| | - Shu-Yuan Xiao
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
| | - Kathleen Bengali
- OncoPlex Diagnostics Inc., Rockville, Maryland, United States of America
| | - Jamar Uzzell
- OncoPlex Diagnostics Inc., Rockville, Maryland, United States of America
| | - Marlene Darfler
- OncoPlex Diagnostics Inc., Rockville, Maryland, United States of America
| | - David B. Krizman
- OncoPlex Diagnostics Inc., Rockville, Maryland, United States of America
| | - Fabiola Cecchi
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Donald P. Bottaro
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Theodore Karrison
- Department of Health Studies, University of Chicago, Chicago, Illinois, United States of America
| | | | - Todd Hembrough
- OncoPlex Diagnostics Inc., Rockville, Maryland, United States of America
| | - Jon Burrows
- OncoPlex Diagnostics Inc., Rockville, Maryland, United States of America
| |
Collapse
|
107
|
Albiges L, Guegan J, Le Formal A, Verkarre V, Rioux-Leclercq N, Sibony M, Bernhard JC, Camparo P, Merabet Z, Molinie V, Allory Y, Orear C, Couvé S, Gad S, Patard JJ, Escudier B. MET is a potential target across all papillary renal cell carcinomas: result from a large molecular study of pRCC with CGH array and matching gene expression array. Clin Cancer Res 2014; 20:3411-21. [PMID: 24658158 DOI: 10.1158/1078-0432.ccr-13-2173] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Papillary renal cell carcinomas (pRCC) are the most common nonclear cell RCC subtype. Germline mutations of the MET oncogene at 7q31 have been detected in patients with hereditary type I pRCC and in 13% of sporadic type I pRCC. Recent report of MET inhibition strengthened the role of c-Met inhibition across pRCC. EXPERIMENTAL DESIGN We collected 220 frozen samples of sporadic pRCC through the French RCC Network and quality controlled for percentage of malignant cells >70%. Gene expression was assessed on 98 pRCC using human whole-genome Agilent 8 × 60K arrays. Copy number alterations were analyzed using Agilent Human 2 × 400K and 4× 180K array for type II pRCC and comparative genomic microarray analysis method for type I pRCC. MET gene sequencing was performed on type I pRCC. RESULTS MET expression level was high across all pRCC. We identified copy number alterations (gain) in 46% of type II pRCC and in 81% of type I pRCC. Correlation between DNA copy number alterations and mRNA expression level was highly significant. Eleven somatic mutations of MET gene were identified amongst 51 type I pRCC (21.6%), including 4 new mutations. We validated LRRK2 cokinase as highly correlated to MET expression. CONCLUSION The present report expands the role of MET activation as a potential target across all pRCC subtypes. These data support investigating MET inhibitors in pRCC in correlation with MET activation status.
Collapse
Affiliation(s)
- Laurence Albiges
- Authors' Affiliations: Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France; INSERM U753, IGR, Villejuif, France;
| | | | | | - Virginie Verkarre
- Department of Pathology, Necker-Enfants Malades Hospital, AP-HP, Université Paris Descartes
| | - Nathalie Rioux-Leclercq
- Department of Pathology, CHU Rennes, Faculté de Médecine, Université de Rennes1, Rennes; and
| | - Mathilde Sibony
- Department of Pathology, Tenon Hospital, AP-HP, Université Paris Pierre et Marie Curie
| | | | | | - Zahira Merabet
- Department of Pathology, Institut Gustave Roussy, Villejuif, France
| | | | - Yves Allory
- Department of Pathology, Hopital Mondor, Faculté Paris Sud, Creteil
| | | | - Sophie Couvé
- INSERM U753, IGR, Villejuif, France; Laboratoire de Génétique Oncologique EPHE, Institut Gustave Roussy
| | - Sophie Gad
- INSERM U753, IGR, Villejuif, France; Laboratoire de Génétique Oncologique EPHE, Institut Gustave Roussy
| | - Jean-Jacques Patard
- INSERM U753, IGR, Villejuif, France; Department of Urology, Kremlin Bicetre Hospital, Université Paris Sud, Kremlin Bicêtre, Paris
| | - Bernard Escudier
- Authors' Affiliations: Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France; INSERM U753, IGR, Villejuif, France
| |
Collapse
|
108
|
Affiliation(s)
- Florian Lordick
- University Cancer Center Leipzig (UCCL), University Clinic Leipzig, D-04103 Leipzig, Germany.
| |
Collapse
|
109
|
Smyth EC, Cunningham D. Operable gastro-oesophageal junctional adenocarcinoma: Where to next? World J Gastrointest Oncol 2014; 6:145-155. [PMID: 24936225 PMCID: PMC4058722 DOI: 10.4251/wjgo.v6.i6.145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/13/2014] [Accepted: 04/11/2014] [Indexed: 02/05/2023] Open
Abstract
Oesophageal junctional adenocarcinoma is a challenging and increasingly common disease. Optimisation of pre-operative staging and consolidation of surgery in large volume centres have improved outcomes, however the preferred adjunctive treatment approach remains a matter of debate. This review examines the benefits of neoadjuvant, peri-operative, and post-operative chemotherapy and chemoradiotherapy in this setting in an attempt to reach an evidence based conclusion. Recent findings relating to the molecular characterisation of oesophagogastric cancer and their impact on therapeutics are explored, in addition to the potential benefits of fluoro-deoxyglucose positron emission tomography (FDG-PET) directed therapy. Finally, efforts to decrease the incidence of junctional adenocarcinoma using early intervention in Barrett’s oesophagus are discussed, including the roles of screening, endoscopic mucosal resection, ablative therapies and chemoprevention.
Collapse
|
110
|
Smyth EC, Sclafani F, Cunningham D. Emerging molecular targets in oncology: clinical potential of MET/hepatocyte growth-factor inhibitors. Onco Targets Ther 2014; 7:1001-14. [PMID: 24959087 PMCID: PMC4061165 DOI: 10.2147/ott.s44941] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The MET/hepatocyte growth-factor (HGF) signaling pathway plays a key role in the processes of embryogenesis, wound healing, and organ regeneration. Aberrant activation of MET/HGF occurs through multiple mechanisms including gene amplification, mutation, protein overexpression, and abnormal gene splicing interrupting autocrine and paracrine regulatory feedback mechanisms. In many cancers including non-small-cell lung cancer, colorectal, gastric, renal, and hepatocellular cancer, dysregulation of MET may lead to a more aggressive cancer phenotype and may be a negative prognostic indicator. Successful therapeutic targeting of the MET/HGF pathway has been achieved using monoclonal antibodies against the MET receptor and its ligand HGF in addition to MET-specific and multitargeted small-molecule tyrosine-kinase inhibitors with several drugs in late-phase clinical trials including onartuzumab, rilotumumab, tivantinib, and cabozantinib. MET frequently interacts with other key oncogenic tyrosine kinases including epidermal growth-factor receptor (EGFR) and HER-3 and these interactions may be responsible for resistance to anti-EGFR therapies. Similarly, resistance to MET inhibition may be mediated through EGFR activation, or alternatively by increasing levels of MET amplification or acquisition of novel "gatekeeper" mutations. In order to optimize development of effective inhibitors of the MET/HGF pathway clinical trials must be enriched for patients with demonstrable MET-pathway dysregulation for which robustly standardized and validated assays are required.
Collapse
Affiliation(s)
- Elizabeth C Smyth
- Department of Gastrointestinal Oncology, Royal Marsden Hospital, Sutton, UK
| | - Francesco Sclafani
- Department of Gastrointestinal Oncology, Royal Marsden Hospital, Sutton, UK
| | - David Cunningham
- Department of Gastrointestinal Oncology, Royal Marsden Hospital, Sutton, UK
| |
Collapse
|
111
|
Yang W, Raufi A, Klempner SJ. Targeted therapy for gastric cancer: molecular pathways and ongoing investigations. Biochim Biophys Acta Rev Cancer 2014; 1846:232-7. [PMID: 24858418 DOI: 10.1016/j.bbcan.2014.05.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/12/2014] [Accepted: 05/16/2014] [Indexed: 02/07/2023]
Abstract
Gastric cancer is currently the second leading cause of worldwide cancer mortality. Ongoing collaborative sequencing efforts have highlighted recurrent somatic genomic aberrations in gastric cancer, however, despite advances in characterizing the genomic landscape, there have been few advances in patient outcomes. Prognosis remains poor with a median overall survival of 12 months for advanced disease. The improved survival with trastuzumab, and more recently ramucirumab, underscore the promise of targeted and biologic therapies and the importance of molecular tumor characterization in gastric cancer. Here we review the most frequent actionable alterations in gastric cancer and highlight ongoing clinical investigations attempting to translate biologic understanding into improved clinical outcomes.
Collapse
Affiliation(s)
- Wei Yang
- University of California Irvine, Department of Medicine, Orange, CA, USA
| | - Alexander Raufi
- University of California Irvine, Department of Medicine, Orange, CA, USA
| | - Samuel J Klempner
- University of California Irvine, Division of Hematology-Oncology, Orange, CA, USA.
| |
Collapse
|
112
|
Aprile G, Giampieri R, Bonotto M, Bittoni A, Ongaro E, Cardellino GG, Graziano F, Giuliani F, Fasola G, Cascinu S, Scartozzi M. The challenge of targeted therapies for gastric cancer patients: the beginning of a long journey. Expert Opin Investig Drugs 2014; 23:925-42. [PMID: 24806575 DOI: 10.1517/13543784.2014.912631] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Despite significant improvements in systemic chemotherapy over the last two decades, the prognosis of patients with advanced disease remains dismal. Collaborative, high-quality research and advances in high-throughput technologies have contributed to elucidate molecular pathways underpinning disease progression and have stimulated many clinical studies testing target therapies in the advanced disease setting. Although progress has been made thanks to trastuzumab in HER2 positive tumours, antiangiogenic drugs have produced conflicting results and EGFR-inhibitors have failed to show major improvements. AREAS COVERED While commenting on the results of many key Phase III randomized trials, the Authors discuss the most promising classes of novel targeted agents and present the current challenges toward a customized treatment. EXPERT OPINION Palliative chemotherapy became the worldwide standard of care for patients with advanced gastric cancers, producing significant life prolongation and improvement of life quality. Nevertheless, long-term outcomes of those patients remain poor. Because of the encouraging advancement in novel targeted therapies, such a disappointing scenario is now evolving. While results serve as a springboard for future research, more comprehensive efforts are needed to clarify the biological mechanisms underpinning cancer progression and help clinicians to develop new effective treatments.
Collapse
Affiliation(s)
- Giuseppe Aprile
- University and General Hospital, Department of Medical Oncology , Piazzale S Maria della Misericordia 1, 33100, Udine , Italy +39 432 559308 ; +39 432 559305 ;
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Mai A, Muharram G, Barrow-McGee R, Baghirov H, Rantala J, Kermorgant S, Ivaska J. Distinct c-Met activation mechanisms induce cell rounding or invasion through pathways involving integrins, RhoA and HIP1. J Cell Sci 2014; 127:1938-52. [DOI: 10.1242/jcs.140657] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
ABSTRACT
Many carcinomas have acquired oncogenic mechanisms for activating c-Met, including c-Met overexpression and excessive autocrine or paracrine stimulation with hepatocyte growth factor (HGF). However, the biological outcome of c-Met activation through these distinct modes remains ambiguous. Here, we report that HGF-mediated c-Met stimulation triggers a mesenchymal-type collective cell invasion. By contrast, the overexpression of c-Met promotes cell rounding. Moreover, in a high-throughput siRNA screen that was performed using a library of siRNAs against putative regulators of integrin activity, we identified RhoA and the clathrin-adapter protein HIP1 as crucial c-Met effectors in these morphological changes. Transient RhoA activation was necessary for the HGF-induced invasion, whereas sustained RhoA activity regulated c-Met-induced cell rounding. In addition, c-Met-induced cell rounding correlated with the phosphorylation of filamin A and the downregulation of active cell-surface integrins. By contrast, a HIP1-mediated increase in β1-integrin turnover was required for the invasion triggered by HGF. Taken together, our results indicate that c-Met induces distinct cell morphology alterations depending on the stimulus that activates c-Met.
Collapse
Affiliation(s)
- Anja Mai
- Turku Centre for Biotechnology, University of Turku, Turku 20521, Finland
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku 20520, Finland
- Department of Biochemistry and Food Chemistry, University of Turku, Turku 20521, Finland
| | - Ghaffar Muharram
- Turku Centre for Biotechnology, University of Turku, Turku 20521, Finland
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku 20520, Finland
| | - Rachel Barrow-McGee
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Habib Baghirov
- Turku Centre for Biotechnology, University of Turku, Turku 20521, Finland
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku 20520, Finland
| | - Juha Rantala
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku 20520, Finland
| | - Stéphanie Kermorgant
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku, Turku 20521, Finland
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku 20520, Finland
- Department of Biochemistry and Food Chemistry, University of Turku, Turku 20521, Finland
| |
Collapse
|
114
|
Abstract
BACKGROUND Gastric cancer is the second most common cause of cancer-related deaths worldwide. There are large geographic variations in the incidence of these tumors, with 60% occurring in East Asia. For patients with resectable disease, surgery and perioperative treatment can be effective. For patients with advanced gastric cancer, chemotherapy regimens result in a median survival of 9-11 months. In general, the prognosis for advanced disease is poor and 5-year overall survival rates are around 15%. Combination therapies yield better survival rates, albeit with increased toxicity. Therefore, more effective and less toxic treatment regimens are needed. SUMMARY The molecular aberrations that characterize the different subgroups of gastric cancer have been used as therapeutic targets. However, the heterogeneity and complexity of gastric cancers is a major challenge for the development of effective targeted therapies. This review examines the main molecular targets in the treatment of gastric cancer, namely the vascular endothelial growth factor (VEGF), human epidermal growth factor receptor 2 (HER2), hepatocyte growth factor (HGF)/c-Met, epidermal growth factor receptor (EGFR) and phosphoinositide 3-kinase (PI3K)/Akt pathways. KEY MESSAGE The molecular aberrations characteristic of gastric cancer are being explored for the development of targeted therapies, including the VEGF, HER2, HGF/c-Met, EGFR and PI3K/Akt signaling pathways. PRACTICAL IMPLICATIONS Trastuzumab, an antibody which targets HER2, is the first approved targeted therapy for the treatment of gastric cancer. However, trastuzumab is only effective in HER2-positive tumors (about 10-20% of all gastric cancers). Ramucirumab, which targets the VEGF receptor 2, has yielded benefits with respect to overall survival in a phase III trial and is an effective treatment for advanced gastric cancer with approval in second-line treatment. Apatinib and rilotumumab are another two promising new agents currently under development.
Collapse
Affiliation(s)
- Nadine Schulte
- Department of Medicine II, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | | | | |
Collapse
|
115
|
Rampa C, Tian E, Våtsveen TK, Buene G, Slørdahl TS, Børset M, Waage A, Sundan A. Identification of the source of elevated hepatocyte growth factor levels in multiple myeloma patients. Biomark Res 2014; 2:8. [PMID: 24716444 PMCID: PMC4022385 DOI: 10.1186/2050-7771-2-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 03/29/2014] [Indexed: 12/20/2022] Open
Abstract
Background Hepatocyte growth factor (HGF) is a pleiotropic cytokine which can lead to cancer cell proliferation, migration and metastasis. In multiple myeloma (MM) patients it is an abundant component of the bone marrow. HGF levels are elevated in 50% of patients and associated with poor prognosis. Here we aim to investigate its source in myeloma. Methods HGF mRNA levels in bone marrow core biopsies from healthy individuals and myeloma patients were quantified by real-time PCR. HGF gene expression profiling in CD138+ cells isolated from bone marrow aspirates of healthy individuals and MM patients was performed by microarray analysis. HGF protein concentrations present in peripheral blood of MM patients were measured by enzyme-linked immunosorbent assay (ELISA). Cytogenetic status of CD138+ cells was determined by fluorescence in situ hybridization (FISH) and DNA sequencing of the HGF gene promoter. HGF secretion in co-cultures of human myeloma cell lines and bone marrow stromal cells was measured by ELISA. Results HGF gene expression profiling in both bone marrow core biopsies and CD138+ cells showed elevated HGF mRNA levels in myeloma patients. HGF mRNA levels in biopsies and in myeloma cells correlated. Quantification of HGF protein levels in serum also correlated with HGF mRNA levels in CD138+ cells from corresponding patients. Cytogenetic analysis showed myeloma cell clones with HGF copy numbers between 1 and 3 copies. There was no correlation between HGF copy number and HGF mRNA levels. Co-cultivation of the human myeloma cell lines ANBL-6 and JJN3 with bone marrow stromal cells or the HS-5 cell line resulted in a significant increase in secreted HGF. Conclusions We here show that in myeloma patients HGF is primarily produced by malignant plasma cells, and that HGF production by these cells might be supported by the bone marrow microenvironment. Considering the fact that elevated HGF serum and plasma levels predict poor prognosis, these findings are of particular importance for patients harbouring a myeloma clone which produces large amounts of HGF.
Collapse
Affiliation(s)
- Christoph Rampa
- The K. G. Jebsen Center for Myeloma Research and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Erming Tian
- The Donna D. and Donald M. Lambert Laboratory of Myeloma Genetics, Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Thea Kristin Våtsveen
- The K. G. Jebsen Center for Myeloma Research and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Glenn Buene
- The K. G. Jebsen Center for Myeloma Research and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tobias Schmidt Slørdahl
- The K. G. Jebsen Center for Myeloma Research and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Magne Børset
- The K. G. Jebsen Center for Myeloma Research and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anders Waage
- The K. G. Jebsen Center for Myeloma Research and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Section of Hematology, St. Olavs University Hospital, Trondheim, Norway
| | - Anders Sundan
- The K. G. Jebsen Center for Myeloma Research and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
116
|
Clinical impact of the HGF/MET pathway activation in patients with advanced gastric cancer treated with palliative chemotherapy. THE PHARMACOGENOMICS JOURNAL 2014; 14:418-23. [DOI: 10.1038/tpj.2014.11] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/17/2014] [Accepted: 01/31/2014] [Indexed: 12/18/2022]
|
117
|
Kluth M, Reynolds K, Rink M, Chun F, Dahlem R, Fisch M, Höppner W, Wagner W, Doh O, Terracciano L, Simon R, Sauter G, Minner S. Reduced membranous MET expression is linked to bladder cancer progression. Cancer Genet 2014; 207:147-52. [PMID: 24853099 DOI: 10.1016/j.cancergen.2014.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 03/15/2014] [Accepted: 03/17/2014] [Indexed: 11/24/2022]
Abstract
The MET protein is involved in the malignant progression of different tumors. This study aimed to analyze the relationship of MET expression with tumor phenotype and clinical outcome in bladder cancer and the role of gene amplification for MET overexpression. A bladder cancer tissue microarray containing 686 bladder cancers was analyzed by immunohistochemistry and by fluorescence in situ hybridization. MET immunostaining was seen in normal urothelium and was recorded in 459 of 560 analyzable urothelial carcinomas (82.0%). Low MET staining was associated with a more unfavorable tumor phenotype. MET staining was seen in 89.8% of 266 pTa, 81.1% of 132 pT1, and 69.4% of 160 pT2-4 cancers (P < 0.0001). MET staining was detectable in 92.4% of 66 grade 1, 85.6% of 257 grade 2, and 75.1% of 237 grade 3 cancers (P = 0.001). MET expression status was not associated with overall or tumor-specific survival in muscle-invasive cancers (pT2-4), tumor progression in pT1 cancers, or recurrences in pTa tumors. Only four of the analyzed tumors (0.8%) showed amplification of the MET gene. We conclude that MET is not overexpressed in urothelial cancer but rather downregulated in a fraction of cancers. Accordingly, rare amplification of the genomic area including the MET gene was not associated with MET protein overexpression.
Collapse
Affiliation(s)
- Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristina Reynolds
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Rink
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Chun
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roland Dahlem
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Margit Fisch
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wolfgang Höppner
- Department of Urology, Clinical Center Itzehoe, Itzehoe, Germany
| | - Walter Wagner
- Department of Urology, German Armed Forces Hospital, Hamburg, Germany
| | - Ousman Doh
- Department of Urology, Regio Clinic Wedel, Wedel, Germany
| | | | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
118
|
Theranostic Profiling for Actionable Aberrations in Advanced High Risk Osteosarcoma with Aggressive Biology Reveals High Molecular Diversity: The Human Fingerprint Hypothesis. Oncoscience 2014; 1:167-179. [PMID: 25126591 PMCID: PMC4128257 DOI: 10.18632/oncoscience.21] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The survival of patients with advanced osteosarcoma is poor with limited therapeutic options. There is an urgent need for new targeted therapies based on biomarkers. Recently, theranostic molecular profiling services for cancer patients by CLIA-certified commercial companies as well as in-house profiling in academic medical centers have expanded exponentially. We evaluated molecular profiles of patients with advanced osteosarcoma whose tumor tissue had been analyzed by one of the following methods: 1. 182-gene next-generation exome sequencing (Foundation Medicine, Boston, MA), 2. Immunohistochemistry (IHC)/PCR-based panel (CARIS Target Now, Irving, Tx), 3.Comparative genome hybridization (Oncopath, San Antonio, TX). 4. Single-gene PCR assays, PTEN IHC (MDACC CLIA), 5. UT Houston morphoproteomics (Houston, TX). The most common actionable aberrations occur in the PI3K/PTEN/mTOR pathway. No patterns in genomic alterations beyond the above are readily identifiable, and suggest both high molecular diversity in osteosarcoma and the need for more analyses to define distinct subgroups of osteosarcoma defined by genomic alterations. Based on our preliminary observations we hypothesize that the biology of aggressive and the metastatic phenotype osteosarcoma at the molecular level is similar to human fingerprints, in that no two tumors are identical. Further large scale analyses of osteosarcoma samples are warranted to test this hypothesis.
Collapse
|
119
|
Abstract
The product of a proto-oncogene, the c-Met protein is a transmembrane receptor tyrosine kinase. Its only known ligand, hepatocyte growth factor/scatter factor, regulates cell growth, motility, migration, invasion, proliferation, and angiogenesis. Dysregulation of c-Met and hepatocyte growth factor have been observed in both clear cell and non-clear cell renal cell carcinomas (RCCs), although only papillary RCCs harbor activating mutations in the MET gene. In clear cell RCC, there is evidence of a direct link between loss of von Hippel-Lindau and up-regulation of c-Met. As in other cancers, high expression of c-Met correlates with worse outcomes in RCC. In vitro and in vivo preclinical RCC models demonstrate cancer control with small molecule and antibodies against c-Met. Given these findings, the c-Met pathway is a logical therapeutic target in RCC, and several agents are in clinical testing with early signs of efficacy.
Collapse
|
120
|
Liu YJ, Shen D, Yin X, Gavine P, Zhang T, Su X, Zhan P, Xu Y, Lv J, Qian J, Liu C, Sun Y, Qian Z, Zhang J, Gu Y, Ni X. HER2, MET and FGFR2 oncogenic driver alterations define distinct molecular segments for targeted therapies in gastric carcinoma. Br J Cancer 2014; 110:1169-78. [PMID: 24518603 PMCID: PMC3950883 DOI: 10.1038/bjc.2014.61] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 12/24/2013] [Accepted: 01/08/2014] [Indexed: 02/06/2023] Open
Abstract
Background: Gastric cancer (GC) is a leading cause of cancer deaths worldwide. Since the approval
of trastuzumab, targeted therapies are emerging as promising treatment options for the
disease. This study aimed to explore the molecular segmentation of several known
therapeutics targets, human epidermal growth factor receptor 2 (HER2), MET and
fibroblast growth factor receptor 2 (FGFR2), within GC using clinically approved or
investigational kits and scoring criteria. Knowledge of how these markers are segmented
in the same cohort of GC patients could improve future clinical trial designs. Methods: Using immunohistochemistry (IHC) and FISH methods, overexpression and amplification of
HER2, FGFR2 and MET were profiled in a cohort of Chinese GC samples. The correlations
between anti-tumour sensitivity and the molecular segments of HER2, MET and FGFR2
alterations were further tested in a panel of GC cell lines and the patient-derived GC
xenograft (PDGCX) model using the targeted inhibitors. Results: Of 172 GC patients, positivity for HER2, MET and FGFR2 alternations was found in 23
(13.4%), 21 (12.2%) and 9 (5.2%) patients, respectively. Positivity
for MET was found in 3 of 23 HER2-positive GC patients. Co-positivity for FGFR2 and MET
was found in 1 GC patient, and amplification of the two genes was found in different
tumour cells. Our study in a panel of GC cell lines showed that in most cell lines,
amplification or high expression of a particular molecular marker was mutually exclusive
and in vitro sensitivity to the targeted agents lapatinib, PD173074 and
crizotinib was only observed in cell lines with the corresponding high expression of the
drugs' target protein. SGC031, an MET-positive PDGCX mouse model, responded to
crizotinib but not to lapatinib or PD173074. Conclusions: Human epidermal growth factor receptor 2, MET and FGFR2 oncogenic driver alterations
(gene amplification and overexpression) occur in three largely distinct molecular
segments in GC. A significant proportion of HER2-negative patients may potentially
benefit from MET- or FGFR2-targeted therapies.
Collapse
Affiliation(s)
- Y J Liu
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - D Shen
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai 200127, China
| | - X Yin
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - P Gavine
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - T Zhang
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - X Su
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - P Zhan
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - Y Xu
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - J Lv
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - J Qian
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - C Liu
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - Y Sun
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - Z Qian
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - J Zhang
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - Y Gu
- Department of Translational Science, Asia & Emerging Markets iMed, AstraZeneca R&D, 199 Liangjing Road, Shanghai 201203, China
| | - X Ni
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai 200127, China
| |
Collapse
|
121
|
Koeppen H, Rost S, Yauch RL. Developing biomarkers to predict benefit from HGF/MET pathway inhibitors. J Pathol 2014; 232:210-8. [PMID: 24105670 DOI: 10.1002/path.4268] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 09/11/2013] [Accepted: 09/15/2013] [Indexed: 12/13/2022]
Abstract
Activation of the MET signalling pathway is critical in regulating multiple cellular processes underlying tumourigenic growth and has represented an attractive target for therapeutic intervention in cancer. Early stage clinical studies of multiple agents targeting this pathway have been undertaken, frequently in unselected patient cohorts with variable results. Promising data in patient subgroups in these studies indicate the need for predictive biomarkers to identify the patients most likely to benefit from these therapies. In this review, we discuss the current knowledge of mechanisms of MET activation, the status of the clinical evaluation of MET-targeted therapies, the associated efforts to identify and validate biomarkers, and the considerations and challenges for potential development of companion diagnostics.
Collapse
Affiliation(s)
- Hartmut Koeppen
- Department of Research Pathology, Genentech, Inc, South San Francisco, CA, USA
| | | | | |
Collapse
|
122
|
Durães C, Almeida GM, Seruca R, Oliveira C, Carneiro F. Biomarkers for gastric cancer: prognostic, predictive or targets of therapy? Virchows Arch 2014; 464:367-78. [DOI: 10.1007/s00428-013-1533-y] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 11/12/2013] [Accepted: 12/23/2013] [Indexed: 12/12/2022]
|
123
|
Rocci A, Gambella M, Aschero S, Baldi I, Trusolino L, Cavallo F, Gay F, Larocca A, Magarotto V, Omedè P, Isaia G, Bertotti A, Liberati AM, Catalano L, De Rosa L, Musto P, Vallone R, Falcone A, Drandi D, Ladetto M, Comoglio PM, Boccadoro M, Palumbo A. MET dysregulation is a hallmark of aggressive disease in multiple myeloma patients. Br J Haematol 2014; 164:841-50. [PMID: 24450886 DOI: 10.1111/bjh.12719] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/02/2013] [Indexed: 11/30/2022]
Abstract
Abnormal activation of MET/HGF (Hepatocyte Growth Factor) pathway has been described in several tumours and increased HGF plasmatic levels have been detected in patients with aggressive multiple myeloma (MM). MET and HGF mRNA expression was investigated in 105 samples of purified plasma cells derived from newly diagnosed MM patients treated with bortezomib-based induction therapy. Gene expression was compared with response to therapy and clinical outcome. MET gene copy number was also evaluated. MET mRNA expression was higher in CD138(+) than in CD138(-) cells (median 76·90 vs. 11·24; P = 0·0009). Low MET mRNA expression characterized patients with better response (complete response or very good partial response) compared to other patients (median 56·10 vs. 134·83; P = 0·0006). After a median follow-up of 50 months, patients with high MET mRNA expression displayed a worse progression-free survival (PFS; P = 0·0029) and overall survival (OS; P = 0·0023) compared to those with low MET mRNA levels. Patients with both high MET mRNA expression and high β2-microglobulin level (>5·5 mg/l) had further worse median PFS (P < 0·0001) and OS (P < 0·0001). Patients carrying 4 MET gene copies (8 out of 82, 9·8%) also had a short PFS. High MET mRNA expression identifies patients with dismal PFS and OS and the combination with high β2-microglobulin further characterizes patients with worse outcome.
Collapse
Affiliation(s)
- Alberto Rocci
- Myeloma Unit, Division of Haematology, University of Torino, Azienda Ospedaliera Città della Salute e della Scienza di Torino, Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Prognostic significance of MET amplification and expression in gastric cancer: a systematic review with meta-analysis. PLoS One 2014; 9:e84502. [PMID: 24416238 PMCID: PMC3885582 DOI: 10.1371/journal.pone.0084502] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 11/21/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND AIMS MET, the hepatocyte growth factor receptor, is a receptor tyrosine kinase overexpressed and activated in a subset of gastric cancer. Several studies investigated the relationship between MET amplification and expression with the clinical outcome in patients with gastric cancer, but yielded conflicting results. We performed a systematic review and meta-analysis to determine the influence of MET amplification and expression on prognosis in gastric cancer. METHODS MEDLINE and EMBASE were searched for studies that explored the association between MET amplification and expression with survival in patients with gastric cancer up to 1 April, 2013. Data of individual hazard ratios (HRs) and 95% confidence intervals (CIs) for meta-analyses were extracted from the publications and combined in pooled HRs. RESULTS Fourteen studies involving 2,258 patients with gastric cancer were included. It was suggested that MET overexpression had an unfavorable impact on survival of patients with gastric cancer, with HRs (95% CIs) of 2.57 (95% CI: 1.97-3.35) overall, 2.82 (95% CI: 1.86-4.27) among studies using amplification for measure scale of MET and 2.42 (95% CI: 1.66-3.54) for expression. The magnitude of association was reduced whereas remained statistically significant in high quality studies or in larger sample size studies and corresponding HRs were 2.18(1.76, 2.70) and 2.35(1.93, 2.87), respectively, without significant heterogeneity. CONCLUSION The findings from present study indicated that higher MET gene amplification and expression in gastric cancer was an indicator of poor prognosis.
Collapse
|
125
|
The OCT4 pseudogene POU5F1B is amplified and promotes an aggressive phenotype in gastric cancer. Oncogene 2013; 34:199-208. [PMID: 24362523 DOI: 10.1038/onc.2013.547] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 11/12/2013] [Accepted: 11/15/2013] [Indexed: 12/15/2022]
Abstract
POU5F1B (POU domain class 5 transcription factor 1B), a processed pseudogene that is highly homologous to OCT4, was recently shown to be transcribed in cancer cells, but its clinical relevance and biological function have remained unclear. We now show that POU5F1B, which is located adjacent to MYC on human chromosome 8q24, is frequently amplified in gastric cancer (GC) cell lines. POU5F1B, but not OCT4, was also found to be expressed at a high level in GC cell lines and clinical specimens. In addition, the DNA copy number and mRNA abundance for POU5F1B showed a positive correlation in both cancer cell lines and GC specimens. Overexpression of POU5F1B in GC cells promoted colony formation in vitro as well as both tumorigenicity and tumor growth in vivo, and these effects were enhanced in the additional presence of MYC overexpression. Furthermore, knockdown of POU5F1B expression with a short hairpin RNA confirmed a role for the endogenous pseudogene in the promotion of cancer cell growth in vitro and tumor growth in vivo. POU5F1B overexpression induced upregulation of various growth factors in GC cells as well as exhibited mitogenic, angiogenic and antiapoptotic effects in GC xenografts. Finally, amplification of POU5F1B was detected in 17 (12%) of 145 cases of GC and was a significant predictor of poor prognosis in patients with stage IV disease. In conclusion, we found that the POU5F1B pseudogene is amplified and expressed at a high level in, as well as confers an aggressive phenotype on, GC, and that POU5F1B amplification is associated with a poor prognosis in GC patients.
Collapse
|
126
|
Chiba S, Tsuchiya N, Horikawa Y, Narita S, Inoue T, Akihama S, Saito M, Numakura K, Tsuruta H, Huang M, Satoh S, Habuchi T. Functional mononucleotide repeat polymorphism in the promoter region of HGF is associated with risk and malignant aggressiveness of bladder cancer. Int J Oncol 2013; 44:678-84. [PMID: 24366484 DOI: 10.3892/ijo.2013.2221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 11/18/2013] [Indexed: 11/05/2022] Open
Abstract
Increased expression of hepatocyte growth factor (HGF) has been shown to be associated with aggressiveness in several types of cancer. Shorter variants of deoxyadenosine tract element (DATE) located in the HGF promoter region have been reported to enhance the expression of HGF. In this study, we investigated the role of HGF DATE variants in bladder cancer risk, HGF expression and clinicopathological features. The frequency of individuals with a short DATE (<28 repeats) in peripheral blood lymphocytes (PBLs) was significantly higher in bladder cancer patients compared to controls (p<0.001). Somatic mutations were observed in 37 of 70 bladder tumor (BT) tissues and the frequency of mutation to long DATE was significantly higher than that to short DATE (p=0.047). The presence of the short DATE in BT tissue was significantly associated with higher tumor grade (p=0.015). HGF mRNA levels were significantly higher in pT2 tumors than pTa or pT1 tumors (p=0.019), and in grade 3 tumors than grade 1 or 2 tumors (p=0.020). Furthermore, BT tissues with the short DATE showed significantly higher levels of HGF mRNA (p<0.001). In patients who underwent radical cystectomy, those with higher HGF expression had a significantly shorter overall survival than those with lower HGF expression (p=0.012). In conclusion, HGF may be associated with the prognosis of patients who undergo radical cystectomy, and the HGF DATE may affect the risk and aggressiveness of bladder cancer by altering HGF expression.
Collapse
Affiliation(s)
- Syuji Chiba
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Norihiko Tsuchiya
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yohei Horikawa
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Shintaro Narita
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Takamitsu Inoue
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Susumu Akihama
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Mitsuru Saito
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Kazuyuki Numakura
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroshi Tsuruta
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Mingguo Huang
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Shigeru Satoh
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Tomonori Habuchi
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
127
|
Cui JJ. Targeting receptor tyrosine kinase MET in cancer: small molecule inhibitors and clinical progress. J Med Chem 2013; 57:4427-53. [PMID: 24320965 DOI: 10.1021/jm401427c] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The HGF/MET signaling pathway is critical in mediating a wide range of normal physiological functions including embryological development, wound healing, and tissue regeneration. Aberrant activation of the pathway has frequently been found in human cancers via protein overexpression, mutation, gene amplification, and also paracrine or autocrine up-regulation. In addition, the activation of HGF/MET signaling confers resistance to the effects of cancer treatments. Therefore, inhibition of the HGF/MET signaling pathway has great potential for therapeutic intervention in cancer. Currently, there are three approaches toward modulating HGF/MET signaling in human clinical studies of cancer: anti-HGF monoclonal antibodies, MET monoclonal antibodies, and small molecule MET inhibitors. Preliminary clinical benefit from inhibition of HGF or MET has been reported. This Perspective will provide an overview of the HGF/MET signaling pathway in cancer and then will review the development of small molecule MET inhibitors and their progress in clinical applications.
Collapse
Affiliation(s)
- J Jean Cui
- TP Therapeutics, Inc. , 6150 Lusk Boulevard, Suite B100, San Diego, California 92121, United States
| |
Collapse
|
128
|
A phase II trial of a selective c-Met inhibitor tivantinib (ARQ 197) monotherapy as a second- or third-line therapy in the patients with metastatic gastric cancer. Invest New Drugs 2013; 32:355-61. [DOI: 10.1007/s10637-013-0057-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 12/01/2013] [Indexed: 12/11/2022]
|
129
|
Ha SY, Lee J, Kang SY, Do IG, Ahn S, Park JO, Kang WK, Choi MG, Sohn TS, Bae JM, Kim S, Kim M, Kim S, Park CK, Ignatius Ou SH, Kim KM. MET overexpression assessed by new interpretation method predicts gene amplification and poor survival in advanced gastric carcinomas. Mod Pathol 2013; 26:1632-41. [PMID: 23807774 DOI: 10.1038/modpathol.2013.108] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/06/2013] [Accepted: 05/11/2013] [Indexed: 12/13/2022]
Abstract
The establishment of better selection criteria for identifying sub-populations that may benefit from treatment is a key aspect of the development and success of targeted therapy. To investigate methods for assessing MET overexpression in gastric cancer, we conducted immunohistochemistry using a new anti-Total MET monoclonal antibody in a single-institution cohort of 495 patients. As antibody is directed against a membranous and/or cytoplasmic epitope, two interpretation methods were used: (1) membranous and cytoplasmic and (2) membranous alone. In selected 120 cases, copy number gain and mRNA expression levels were measured using quantitative real-time PCR. Further in situ hybridization confirmed the presence of MET gene amplification. Among the 495 gastric cancers, simultaneous membranous and cytoplasmic overexpression of MET was found in 108 cases (21.8%) and membranous alone overexpression was observed in 40 cases (8.1%). The highest correlation was observed in membranous and cytoplasmic staining of MET: MET expression scores correlated significantly with high MET mRNA levels (r=0.465, P<0.0001), increased copy number gain (r=0.393, P=0.000002) and amplification of MET gene. Moreover, patients with MET overexpression showed shorter overall survival (HR, 1.781; 95% CI, 1.324-2.395; P<0.001) and disease-free survival (HR, 1.765; 95% CI, 1.227-2.541; P=0.002) compared with patients without MET overexpression. However, membranous overexpression of MET did not highly correlate with mRNA level (r=0.274, P=0.002), copy number gain or survival (P>0.05). We developed highly correlating interpretation methods of MET immunohistochemistry in gastric carcinomas. MET overexpression is an independent prognostic factor and could be a potential target and predictor of benefit for targeted therapy with MET inhibitors.
Collapse
Affiliation(s)
- Sang Y Ha
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Zhao L, Yasumoto K, Kawashima A, Nakagawa T, Takeuchi S, Yamada T, Matsumoto K, Yonekura K, Yoshie O, Yano S. Paracrine activation of MET promotes peritoneal carcinomatosis in scirrhous gastric cancer. Cancer Sci 2013; 104:1640-6. [PMID: 24118504 DOI: 10.1111/cas.12301] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 10/01/2013] [Accepted: 10/07/2013] [Indexed: 12/17/2022] Open
Abstract
Scirrhous gastric cancer is associated with abundant stroma and frequently develops into peritoneal carcinomatosis with malignant ascites. Although malignant ascites is among the most deadly diseases worldwide, its molecular pathogenesis is poorly understood. We investigated the role of hepatocyte growth factor (HGF) in the production of peritoneal carcinomatosis with malignant ascites. We examined three scirrhous and three non-scirrhous human gastric cancer cell lines for the production of peritoneal carcinomatosis in vivo and responses to HGF in vitro. Furthermore, clinical scirrhous gastric cancer specimens were examined for HGF production. Among the six cell lines examined, only two scirrhous cell lines (NUGC4 and GCIY) produced peritoneal carcinomatosis with massive ascites after intraperitoneal injection in nude mice. Their proliferation was stimulated by exogenous HGF in vitro. On the other hand, a non-scirrhous cell line, MKN45, with MET amplification generated peritoneal tumors but not ascites. MET tyrosine kinase inhibitors, crizotinib and TAS-115, inhibited HGF-stimulated proliferation of NUGC4 and GCIY as well as constitutive proliferation of MKN45. Furthermore, crizotinib and TAS-115 prolonged the survival of mice bearing established tumors by NUGC4 or MKN45. In clinical specimens, HGF was markedly produced by stromal fibroblasts. Malignant ascitic fluids from patients with peritoneal carcinomatosis contained high levels of HGF. Our results strongly suggest that paracrine HGF-induced activation of MET-mediated signaling pathways plays an important role in the pathogenesis of peritoneal carcinomatosis in scirrhous gastric cancer. Thus, MET signaling pathway may be a potential therapeutic target for peritoneal carcinomatosis of gastric cancer, even without MET amplification.
Collapse
Affiliation(s)
- Lu Zhao
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
An X, Wang F, Shao Q, Wang FH, Wang ZQ, Wang ZQ, Chen C, Li C, Luo HY, Zhang DS, Xu RH, Li YH. MET amplification is not rare and predicts unfavorable clinical outcomes in patients with recurrent/metastatic gastric cancer after chemotherapy. Cancer 2013; 120:675-82. [PMID: 24804300 DOI: 10.1002/cncr.28454] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Several large studies have reported an extremely low incidence of MET gene amplification (GA) in patients with radically resected gastric cancer. This study was conducted to evaluate the prevalence and prognostic role of MET in patients with recurrent=metastatic gastric cancer who received chemotherapy. METHODS MET GA and protein expression of recurrent=metastatic gastric cancer samples were evaluated by fluorescence in situ hybridization and immunohistochemistry (IHC), respectively. RESULTS This retrospective study included 232 patients with recurrent=metastatic gastric cancer. MET GA and strong protein expression(IHC31) were observed in 8.3% (19 of 230 samples) and 9.6% (22 of 229 samples) of samples, respectively. A significant correlation was observed between MET GA and protein expression (r = 0.378; P<.001). MET GA was correlated with poor performance status(P<.001) and poorly differentiated tumors (P=.0015). Both MET GA and IHC 31 expression were associated with a substantially shorter median overall survival (OS) and progression-free survival (PFS). The median OS and PFS for patients with MET GA versus those without MET GA were 5.7 months versus 15.5 months (P<.001) and 3.6 months versus 6.9 months (P<.001), respectively. The median OS and PFS for patients with MET IHC 31 expression versus IHC 0 to 21 expression were 6.3 months versus 15.1 months(P<.001) and 3.6 months versus 7.0 months (P<.001), respectively. CONCLUSIONS In patients with recurrent=metastatic gastric cancer,MET amplification and strong protein expression are not rare and appear to be significantly associated with unfavorable clinical outcomes.
Collapse
Affiliation(s)
- Xin An
- State Key Laboratory of Oncology in South China, Guangzhou, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Yu S, Yu Y, Zhao N, Cui J, Li W, Liu T. C-Met as a prognostic marker in gastric cancer: a systematic review and meta-analysis. PLoS One 2013; 8:e79137. [PMID: 24223894 PMCID: PMC3817069 DOI: 10.1371/journal.pone.0079137] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/17/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND c-Met has been recognized as an important therapeutic target in gastric cancer, but the prognostic property of the c-Met status is still unclear. We aimed to characterize the prognostic effect of c-Met by systematic review and meta-analysis. METHODS We identified 15 studies assessing survival in gastric cancer by c-Met status. Effect measure of interest was hazard ratio (HR) for survival. Meta-regression was performed to estimate the relationship between HR and disease stage. Random-effects meta-analyses were used to account for heterogeneity. RESULTS 15 eligible studies provided outcome data stratified by c-Met status in 2210 patients. Meta-analysis of the HRs indicated a significantly poorer Os in patients with high c-Met expression (average HR=2.112, 95%CI: 1.622-2.748). Subgroup analysis showed the prognostic effect of c-Met was identical in protein-level and gene-level based methodology. The same effect was also seen in Asian and Western ethnicity subgroup analysis. Meta-regression showed HR was not associated with disease stage. CONCLUSIONS Patients with tumors that harbor high c-Met expression are more likely to have a worse Os, with this prognostic effect independent of disease stage. c-Met status should be evaluated in clinical prognosis.
Collapse
Affiliation(s)
- Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Naiqing Zhao
- Department of Biostatistics, Fudan University, Shanghai, People’s Republic of China
| | - Jianlan Cui
- Department of Biostatistics, Fudan University, Shanghai, People’s Republic of China
| | - Wei Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
133
|
Teng L, Lu J. cMET as a potential therapeutic target in gastric cancer (Review). Int J Mol Med 2013; 32:1247-54. [PMID: 24141315 DOI: 10.3892/ijmm.2013.1531] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 10/16/2013] [Indexed: 11/05/2022] Open
Abstract
Gastric cancer is one of the most common malignancies worldwide. Despite improvements in surgery and chemotherapy, the outcomes in patients with advanced gastric cancer remain poor. cMET is a member of the receptor tyrosine kinase family, and plays a key role in tumor survival, growth, angiogenesis and metastasis. cMET overexpression and/or gene amplification occurs in a significant proportion of gastric cancers. cMET is associated with a high tumor stage and poor prognosis. Several cMET inhibitors have been investigated in clinical trials, and the initial results are encouraging. It has become increasingly apparent that cMET is a promising therapeutic target in gastric cancer. In this review, we summarize the development of cMET inhibitors in the preclinical and clinical environment. In addition, we discuss the challenges of cMET-targeted therapy in gastric cancer and explore possible solutions.
Collapse
Affiliation(s)
- Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | | |
Collapse
|
134
|
A c-Met inhibitor increases the chemosensitivity of cancer stem cells to the irinotecan in gastric carcinoma. Br J Cancer 2013; 109:2619-28. [PMID: 24129235 PMCID: PMC3833223 DOI: 10.1038/bjc.2013.638] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 09/19/2013] [Accepted: 09/24/2013] [Indexed: 12/12/2022] Open
Abstract
Background: Cancer stem cells (CSCs) may be postulated mediators of the chemoresistance. This study aimed to determine an effective signal inhibitor with effects on the proliferation of CSCs in combination with anticancer drugs. Methods: We used three gastric cancer cell lines and three side population (SP)-enriched CSC cell lines. We examined the combined effects of inhibitors against stemness signals, including c-Met inhibitor SU11274, and five anticancer drugs on the CSC proliferation and mRNA expression of chemoresistance-associated genes. Results: The IC50 of irinotecan in SP-enriched CSC was 10.5 times higher than parent OCUM-2M cells, whereas that of oxaliplatin, taxol, gemcitabine, and 5-fluorouracil was 2.0, 2.8, 2.0, and 1.2, respectively. The SP cell lines had higher expression levels of UGT1A1, ABCG2, and ABCB1 than their parent cell lines. There was a synergistic antiproliferative effect with a combination of SU11274 and SN38 in SP cells, but not other inhibitors. The SU11274 significantly decreased the expression of UGT1A1, but not ABCG2 and ABCB1. The SN38 plus SU11274 group more effectively suppressed in vivo tumour growth by OCUM-2M/SP cells than either group alone. Conclusion: Cancer stem cells have chemoresistance to irinotecan. The c-Met inhibitor may be a promising target molecule for irinotecan-based chemotherapy of gastric cancer.
Collapse
|
135
|
Spigel DR, Ervin TJ, Ramlau RA, Daniel DB, Goldschmidt JH, Blumenschein GR, Krzakowski MJ, Robinet G, Godbert B, Barlesi F, Govindan R, Patel T, Orlov SV, Wertheim MS, Yu W, Zha J, Yauch RL, Patel PH, Phan SC, Peterson AC. Randomized phase II trial of Onartuzumab in combination with erlotinib in patients with advanced non-small-cell lung cancer. J Clin Oncol 2013; 31:4105-14. [PMID: 24101053 DOI: 10.1200/jco.2012.47.4189] [Citation(s) in RCA: 388] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Increased hepatocyte growth factor/MET signaling is associated with poor prognosis and acquired resistance to epidermal growth factor receptor (EGFR) -targeted drugs in patients with non-small-cell lung cancer (NSCLC). We investigated whether dual inhibition of MET/EGFR results in clinical benefit in patients with NSCLC. PATIENTS AND METHODS Patients with recurrent NSCLC were randomly assigned at a ratio of one to one to receive onartuzumab plus erlotinib or placebo plus erlotinib; crossover was allowed at progression. Tumor tissue was required to assess MET status by immunohistochemistry (IHC). Coprimary end points were progression-free survival (PFS) in the intent-to-treat (ITT) and MET-positive (MET IHC diagnostic positive) populations; additional end points included overall survival (OS), objective response rate, and safety. RESULTS There was no improvement in PFS or OS in the ITT population (n = 137; PFS hazard ratio [HR], 1.09; P = .69; OS HR, 0.80; P = .34). MET-positive patients (n = 66) treated with erlotinib plus onartuzumab showed improvement in both PFS (HR, .53; P = .04) and OS (HR, .37; P = .002). Conversely, clinical outcomes were worse in MET-negative patients treated with onartuzumab plus erlotinib (n = 62; PFS HR, 1.82; P = .05; OS HR, 1.78; P = .16). MET-positive control patients had worse outcomes versus MET-negative control patients (n = 62; PFS HR, 1.71; P = .06; OS HR, 2.61; P = .004). Incidence of peripheral edema was increased in onartuzumab-treated patients. CONCLUSION Onartuzumab plus erlotinib was associated with improved PFS and OS in the MET-positive population. These results combined with the worse outcomes observed in MET-negative patients treated with onartuzumab highlight the importance of diagnostic testing in drug development.
Collapse
Affiliation(s)
- David R Spigel
- David R. Spigel, Thomas J. Ervin, and Davey B. Daniel, Sarah Cannon Research Institute; David R. Spigel, Tennessee Oncology, Nashville; Davey B. Daniel, Chattanooga Oncology Hematology Associates, Chattanooga, TN; Thomas J. Ervin, Florida Cancer Specialists, Fort Myers; Michael S. Wertheim, Hematology/Oncology Associates, Port St Lucie, FL; Rodryg A. Ramlau, Poznan University of Medical Sciences, Poznan; Maciej J. Krzakowski, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jerome H. Goldschmidt Jr, Blue Ridge Cancer Care, Christianburg, VA; George R. Blumenschein Jr, The University of Texas MD Anderson Cancer Center, Houston, TX; Gilles Robinet, University Hospital Morvan, Brest; Benoit Godbert, Centre Hospitalier Universitaire Nancy, Vandoeuvre-lès-Nancy; Fabrice Barlesi, Assistance Publique-Hôpitaux de Marseille, Aix Marseille University, Marseille, France; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; Taral Patel, Mid Ohio Oncology/Hematology, Columbus, OH; Sergey V. Orlov, St Petersburg Pavlov State Medical University, St Petersburg, Russia; Wei Yu, Robert L. Yauch, Premal H. Patel, and See-Chun Phan, Genentech; Amy C. Peterson, Medivation, San Francisco, CA; and Jiping Zha, Crown Bioscience, Taicang City, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Knockdown of RON receptor kinase delays but does not prevent tumor progression while enhancing HGF/MET signaling in pancreatic cancer cell lines. Oncogenesis 2013; 2:e76. [PMID: 24100611 PMCID: PMC3816215 DOI: 10.1038/oncsis.2013.36] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 06/07/2013] [Accepted: 08/13/2013] [Indexed: 12/14/2022] Open
Abstract
In this study, the role of RON (receptor originated from nantes) in tumor progression was further investigated in context with MET expression and activity. RON and MET expressions were not detected in an immortalized normal human pancreas cell line (HPNE), but were co-expressed in five of seven pancreatic ductal adenocarcinoma (PDAC) cell lines (PANC-1, BxPC-3, Capan-2, CFPAC-1 and AsPC-1). RON expression was knocked down by an shRNA approach in two PDAC cell lines (BxPC-3 and CFPAC-1) that co-express MET. Knockdown of RON significantly inhibited cell growth, clonogenicity and macrophage stimulating protein (MSP), RON ligand induced invasion by in vitro assays and significantly inhibited tumor growth (P<0.001) and metastasis (P<0.009) in an orthotopic pancreatic cancer mouse model at week 7. However, by week 9, the mice implanted with RON knockdown cells had developed similar size primary tumors and metastases compared with that seen in the control group at week 7. Western blotting and immunohistochemistry analyses showed that MET remains highly expressed in cells and tumor tissues where RON was knocked down. Moreover, knockdown of RON did not prevent hepatocyte growth factor (HGF) stimulated invasion in in vitro Matrigel assays. Treating cells with MSP induced the transphosphorylation of MET, suggesting that signaling may be modulated by relative levels of RON and MET receptors and their corresponding ligands. To this point, HGF treatment of RON knockdown cells caused an increase in intensity and duration of MET signaling, suggesting that MET signaling may compensate for loss of RON signaling. Treatment of cells with an MET inhibitor, PHA-665752, had minimal effects on inhibiting cell growth but significantly inhibited cell invasion induce by ligands for either MET or RON. These results suggest that HGF/MET signaling may have a more important role in tumor cell invasion and metastasis rather than in tumor cell proliferation. This study indicates that specific inhibition of RON delays but does not prevent progression of PDAC. Moreover, specific signaling may be modulated by the interaction of RON and MET receptors. This dynamic interaction of RON and MET in pancreatic cancer cells suggests that dual targeting of both RON and MET will be preferable to inhibition of either target alone.
Collapse
|
137
|
Kawakami H, Okamoto I, Arao T, Okamoto W, Matsumoto K, Taniguchi H, Kuwata K, Yamaguchi H, Nishio K, Nakagawa K, Yamada Y. MET amplification as a potential therapeutic target in gastric cancer. Oncotarget 2013; 4:9-17. [PMID: 23327903 PMCID: PMC3702203 DOI: 10.18632/oncotarget.718] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Our aim was to investigate both the prevalence of MET amplification in gastric cancer as well as the potential of this genetic alteration to serve as a therapeutic target in gastric cancer. MET amplification was assessed by initial screening with a PCR-based copy number assay followed by confirmatory FISH analysis in formalin-fixed, paraffin-embedded specimens of gastric cancer obtained at surgery. The effects of MET tyrosine kinase inhibitors (MET-TKIs) in gastric cancer cells with or without MET amplification were also examined. The median MET copy number in 266 cases of gastric cancer was 1.7, with a range of 0.41 to 21.3. We performed FISH analysis for the 15 cases with the highest MET copy numbers. MET amplification was confirmed in the four assessable cases with a MET copy number of at least 4, whereas MET amplification was not detected in those with a gene copy number of less than 4. The prevalence of MET amplification was thus 1.5% (4 out of 266 cases). Inhibition of MET by MET-TKIs resulted in the induction of apoptosis accompanied by attenuation of downstream MET signaling in gastric cancer cell lines with MET amplification but not in those without this genetic change. MET amplification identifies a small but clinically important subgroup of gastric cancer patients who are likely to respond to MET-TKIs. Furthermore, screening with a PCR-based copy number assay is an efficient way to reduce the number of patients requiring confirmation of MET amplification by FISH analysis.
Collapse
Affiliation(s)
- Hisato Kawakami
- Department of Medical Oncology, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Richards D, Kocs DM, Spira AI, David McCollum A, Diab S, Hecker LI, Cohn A, Zhan F, Asmar L. Results of docetaxel plus oxaliplatin (DOCOX) ± cetuximab in patients with metastatic gastric and/or gastroesophageal junction adenocarcinoma: results of a randomised Phase 2 study. Eur J Cancer 2013; 49:2823-31. [PMID: 23747051 DOI: 10.1016/j.ejca.2013.04.022] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 04/19/2013] [Accepted: 04/25/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Patients with advanced adenocarcinoma of the gastroesophageal junction/stomach are treated by combination chemotherapy, with minimal improvements in survival. We evaluated adding cetuximab to combination chemotherapy in these patients. METHODS The primary objective was progression-free survival. Secondary objectives were response rate, time to response, duration of response and safety. Treatment Arm 1: docetaxel+oxaliplatin (DOCOX)=docetaxel 60 mg/m(2) plus oxaliplatin 130 mg/m(2) on Day 1 of each 21-day cycle. Arm 2: docetaxel+oxaliplatin+cetuximab (DOCOX+C)=DOCOX with C 400mg/m(2) first dose then 250 mg/m(2) weekly. The protocol was amended to allow collection of tissue to correlate responses with KRAS status. FINDINGS One hundred fifty patients were enrolled (75/arm). DOCOX/DOCOX+C: gastric 44%/41%, gastroesophageal junction 51%/55%, both 5%/4%. Response rate/arm: 26.5%/38.0%. Median progression-free survival: 4.7/5.1 months (95% confidence interval (CI) 3.0-5.6/4.3-5.9); 1 year survival: 39.1%/33.0%, median overall survival: 8.5/9.4 months; median duration of response: 7.3/5.6months. Grade 3-4 treatment-related adverse events (%) included neutropenia (50%/44%), febrile neutropenia (13%/19%), diarrhoea (12%/17%), fatigue (12%/17%) and leukopenia (7%/14%). Discontinuation was due to progressive disease 39/32 and adverse events 21/34. KRAS was collected on some patients 2 years into the study because of new American Society of Clinical Oncology (ASCO) findings. INTERPRETATION Cetuximab added to DOCOX may improve response rate minimally; there appears to be no improvement in progression-free survival, overall survival or 1-year survival. Cetuximab added to DOCOX did not produce clinically significant outcomes. Toxicities were consistent with the study drugs' known safety profiles. KRAS mutation was infrequent; no conclusions can be drawn from KRAS response data. ClinicalTrial.gov Identifier: NCT00517829.
Collapse
Affiliation(s)
- Donald Richards
- US Oncology Research, McKesson Specialty Health, The Woodlands, TX, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Abstract
Gastric cancer (GC) is a major health burden throughout the world, especially in certain endemic regions. GC is commonly diagnosed at an advanced stage because of the lack of early detection strategies and is usually associated with a dismal outcome. For patients with localized GC (LGC), surgery is the best cure: cure rates are highly associated with the surgical pathology stage. Adjunctive therapies improve the cure rates by about an additional 10%. Therefore, a multimodality approach is highly recommended for all patients with LGC. This article highlights some of the therapeutic advances made against GC and features important ongoing trials.
Collapse
|
140
|
C-MET is expressed in the majority of penile squamous cell carcinomas and correlates with polysomy-7 but is not associated with MET oncogene amplification, pertinent histopathologic parameters, or with cancer-specific survival. Pathol Res Pract 2013; 209:215-20. [PMID: 23490239 DOI: 10.1016/j.prp.2013.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 11/29/2012] [Accepted: 02/05/2013] [Indexed: 01/02/2023]
Abstract
We assessed c-MET expression and oncogene amplification in a cohort enrolling 92 surgically treated penile squamous cell carcinomas (PSCCs). A tissue microarray was constructed for c-MET immunohistochemistry (IHC) and chromogenic silver in situ hybridization (SISH). Two independent pathologists evaluated IHC by employing the breast cancer scoring rules, and scored the presence of MET oncogene amplification and/or polysomy-7. Eighty study cases (87%) showed c-MET expression. No study case had MET oncogene amplification, but 42 patients (45.7%) had polysomy-7. Polysomy-7 showed a significant positive correlation with c-MET expression (ρ=0.323, p=0.002). Neither c-MET expression nor polysomy-7 was associated with histopathologic parameters or with cancer-specific survival (median post-surgical follow-up 32 months). Our data suggest that the majority of PSCCs exhibit c-MET expression which is not associated with oncogene amplification, but might be attributable to polysomy-7. Further studies should investigate the expression and activation of downstream molecules functionally involved in c-MET pathway signaling, and clarify the so far unresolved role of c-MET inhibitors as potential targeted therapies in PSCCs with metastatic dissemination.
Collapse
|
141
|
Scagliotti GV, Novello S, von Pawel J. The emerging role of MET/HGF inhibitors in oncology. Cancer Treat Rev 2013; 39:793-801. [PMID: 23453860 DOI: 10.1016/j.ctrv.2013.02.001] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/31/2013] [Accepted: 02/02/2013] [Indexed: 02/06/2023]
Abstract
The N-methyl-N'-nitroso-guanidine human osteosarcoma transforming gene (MET) receptor tyrosine kinase and its ligand hepatocyte growth factor (HGF) control cellular signaling cascades that direct cell growth, proliferation, survival, and motility. Aberrant MET/HGF activation has been observed in many tumor types, can occur by multiple mechanisms, and promotes cellular proliferation and metastasis via growth factor receptors and other oncogenic receptor pathways. Thus, MET/HGF inhibition has emerged as targeted anticancer therapies. Preclinically, neoplastic and metastatic phenotypes of several tumor cells, including non-small cell lung cancer, hepatocellular carcinoma, and gastric cancer, were abrogated by MET inhibition. Ongoing clinical development with tivantinib, cabozantinib, onartuzumab, crizotinib, rilotumumab, and ficlatuzumab has shown encouraging results. These trials have established a key role for MET in a variety of tumor types. Evidence is emerging for identification of aberrant MET activity biomarkers and selection of patient subpopulations that may benefit from targeted MET and HGF inhibitor treatment.
Collapse
|
142
|
Huang WT, Chuang SS. High MET gene copy number predicted poor prognosis in primary intestinal diffuse large B-cell lymphoma. Diagn Pathol 2013; 8:16. [PMID: 23379953 PMCID: PMC3584722 DOI: 10.1186/1746-1596-8-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 01/31/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND MET is a proto-oncogene with its copy number (CN) alterations been reported in some cancers, but not in primary intestinal diffuse large B-cell lymphoma (PI-DLBL) yet. METHODS In this retrospective study, we performed histology and chart reviews, immunohistochemistry and quantitative polymerase chain reaction for MET CN alterations on 28 surgically resected PI-DLBLs. RESULTS There were 12 men and 16 women with a median age of 70 and a mean follow-up of 32 months. The median MET CN was 2.20 (range, 1.04 to 3.35). CN gain was observed in 11 cases, including 5 with CN greater than 3. Nine patients (32%) had diploid CN and eight (29%) with CN loss. Patients with gain or diploid CN showed significantly worse prognosis (P = 0.046) than those with CN loss. Furthermore, MET CN greater than 3 was associated with an adverse outcome (P = 0.003). Intestinal perforation at presentation was the sole clinicopathological factor associated with a poor prognosis (P = 0.004) and perforation was correlated with CN greater than 3 (P = 0.002). CONCLUSIONS Our finding of MET CN gain as a poor prognostic factor in PI-DLBL patients might serve as the rationale for targeting MET signaling pathway in the treatment of these patients.
Collapse
Affiliation(s)
- Wan-Ting Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | |
Collapse
|
143
|
Cheung DY, Kim JK. Perspectives of the Stomach Cancer Treatment: The Introduction of Molecular Targeted Therapy and the Hope for Cure. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2013; 61:117-27. [DOI: 10.4166/kjg.2013.61.3.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Dae Young Cheung
- Department of Internal Medicine, The Catholic University of Korea, College of Medicine, Seoul, Korea
| | - Jae Kwang Kim
- Department of Internal Medicine, The Catholic University of Korea, College of Medicine, Seoul, Korea
| |
Collapse
|
144
|
Minuti G, Cappuzzo F, Duchnowska R, Jassem J, Fabi A, O’Brien T, Mendoza AD, Landi L, Biernat W, Czartoryska-Arłukowicz B, Jankowski T, Zuziak D, Zok J, Szostakiewicz B, Foszczyńska-Kłoda M, Tempińska-Szałach A, Rossi E, Varella-Garcia M. Increased MET and HGF gene copy numbers are associated with trastuzumab failure in HER2-positive metastatic breast cancer. Br J Cancer 2012; 107:793-9. [PMID: 22850551 PMCID: PMC3425981 DOI: 10.1038/bjc.2012.335] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 06/28/2012] [Accepted: 07/06/2012] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND To investigate whether copy number gain of MET or hepatocyte growth factor (HGF) affect trastuzumab sensitivity in HER2-positive metastatic breast cancer (MBC). METHODS We analysed 130 HER2-positive MBC treated with trastuzumab-based therapy. MET and HGF gene copy numbers (GCN) were assessed by fluorescence in situ hybridisation (FISH) in primary breast cancer samples. Receiver operating characteristic analysis was applied to find the best cutoff point for both MET and HGF GCN. RESULTS MET FISH-positive cases (N=36, mean 3.72) had a significantly higher trastuzumab failure rate (44.4% vs 16.0%; P=0.001) and a significantly shorter time to progression (5.7 vs 9.9 months; HR 1.74; P=0.006) than MET FISH-negative cases (N=94, mean <3.72). Hepatocyte growth factor GCN was evaluated in 84 cases (64.6%). Receiver operating characteristic analysis identified 33 HGF FISH-positive patients (mean HGF GCN 3.01). HGF FISH-positive status was significantly associated with higher risk of failure (30.3% vs 7.8%; P=0.007) as compared with HGF FISH-negative cases (N=51, mean <3.01). MET and HGF FISH-positive status was highly correlated (P<0.001) and combination of both biomarkers did not increase predictive value of either considered separately. CONCLUSION High GCNs of MET and HGF associate with an increased risk of trastuzumab-based therapy failure in HER2-positive MBC.
Collapse
Affiliation(s)
- G Minuti
- Department of Medical Oncology, Civil Hospital of Livorno, Istituto Toscano Tumori, Viale Alfieri 36, Livorno 57100, Italy
| | - F Cappuzzo
- Department of Medical Oncology, Civil Hospital of Livorno, Istituto Toscano Tumori, Viale Alfieri 36, Livorno 57100, Italy
| | - R Duchnowska
- Department of Oncology, Military Institute of Medicine, 128 Szaserów Street, Warsaw 00-909, Poland
| | - J Jassem
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, 3a M Skłodowskiej-Curie Street, Gdańsk 80-210, Poland
| | - A Fabi
- Department of Medical Oncology, National Cancer Institute Regina Elena, Via E Chianesi 53, RM 00144, Rome, Italy
| | - T O’Brien
- Molecular Pathology Shared Resource, University of Colorado Cancer Center, 13001 E17th Place, CO 80045, Aurora, Colorado, USA
| | - A D Mendoza
- Molecular Pathology Shared Resource, University of Colorado Cancer Center, 13001 E17th Place, CO 80045, Aurora, Colorado, USA
| | - L Landi
- Department of Medical Oncology, Civil Hospital of Livorno, Istituto Toscano Tumori, Viale Alfieri 36, Livorno 57100, Italy
| | - W Biernat
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, 3a M Skłodowskiej-Curie Street, Gdańsk 80-210, Poland
| | - B Czartoryska-Arłukowicz
- Department of Oncology, Białystock Oncology Center, 47 Fabryczna Street, Bialystok 15-047, Poland
| | - T Jankowski
- Department of Oncology, Lublin Oncology Center, 7 Jaczewskiego Street, Lublin PL- 20090, Poland
| | - D Zuziak
- Department of Oncology, Beskidy Oncology Center, 18 Liberation Street, Bielsko-Biała 43-300, Poland
| | - J Zok
- Department of Oncology, Warmia and Masuria Oncology Center, 5 Oczapowskiego Street, Olsztyn 10-967, Poland
| | - B Szostakiewicz
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, 3a M Skłodowskiej-Curie Street, Gdańsk 80-210, Poland
| | - M Foszczyńska-Kłoda
- Department of Oncology, West Pomeranian Oncology Center, 55 Adama Mickiewicza Street, Szczecin 70-385, Poland
| | - A Tempińska-Szałach
- Department of Oncology, District Hospital of Elbląg, 35 Komeńskiego Street, Elbląg 82-300, Poland
| | - E Rossi
- Department of Medical Oncology, Civil Hospital of Livorno, Istituto Toscano Tumori, Viale Alfieri 36, Livorno 57100, Italy
| | - M Varella-Garcia
- Molecular Pathology Shared Resource, University of Colorado Cancer Center, 13001 E17th Place, CO 80045, Aurora, Colorado, USA
| |
Collapse
|
145
|
Pierscianek D, Kim YH, Motomura K, Mittelbronn M, Paulus W, Brokinkel B, Keyvani K, Wrede K, Nakazato Y, Tanaka Y, Mariani L, Vital A, Sure U, Ohgaki H. MET gain in diffuse astrocytomas is associated with poorer outcome. Brain Pathol 2012; 23:13-8. [PMID: 22672415 DOI: 10.1111/j.1750-3639.2012.00609.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 05/11/2012] [Indexed: 12/01/2022] Open
Abstract
Glioblastoma may develop rapidly without evidence for precursor lesions (primary glioblastomas), or progress from diffuse or anaplastic astrocytomas (secondary glioblastomas). Despite having distinct genetic profiles, these glioblastoma subtypes have similar histological features. We hypothesized that the highly malignant phenotype of glioblastoma may be attributable to genetic alterations that are common to both glioblastoma subtypes. In the present study, we first searched for commonly (>35%) amplified genes in glioblastomas with IDH1 mutation (a hallmark of secondary glioblastoma) and those without IDH1 mutation (typical for primary glioblastoma) in data from The Cancer Genome Atlas (TCGA). A total of 25 genes were identified, of which 21 were located at 7q31-34. We then screened 264 gliomas (70 glioblastomas, 112 diffuse astrocytomas, 82 oligodendrogliomas) for gain of the MET at 7q31.2 with quantitative polymerase chain reaction (PCR). MET gain was detected in primary glioblastomas (47%) and secondary glioblastomas (44%), suggesting that this genetic alteration plays a role in the pathogenesis of both glioblastoma subtypes. MET gain was also common in diffuse astrocytomas (38%), but less frequent in oligodendrogliomas (16%). MET gain in diffuse astrocytomas was associated with shorter survival (median, 43.0 vs. 70.7 months; P = 0.004), suggesting that MET gain is a useful prognostic marker for diffuse astrocytomas.
Collapse
Affiliation(s)
- Daniela Pierscianek
- Molecular Pathology, International Agency for Research on Cancer, Lyon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Liu L, Wu N, Li J. Novel targeted agents for gastric cancer. J Hematol Oncol 2012; 5:31. [PMID: 22709792 PMCID: PMC3411478 DOI: 10.1186/1756-8722-5-31] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 06/18/2012] [Indexed: 12/16/2022] Open
Abstract
Contemporary advancements have had little impact on the treatment of gastric cancer (GC), the world’s second highest cause of cancer death. Agents targeting human epidermal growth factor receptor mediated pathways have been a common topic of contemporary cancer research, including monoclonal antibodies (mAbs) and receptor tyrosine kinase inhibitors (TKIs). Trastuzumab is the first target agent evidencing improvements in overall survival in HER2-positive (human epidermal growth factor receptor 2) gastric cancer patients. Agents targeting vascular epithelial growth factor (VEGF), mammalian target of rapamycin (mTOR), and other biological pathways are also undergoing clinical trials, with some marginally positive results. Effective targeted therapy requires patient selection based on predictive molecular biomarkers. Most phase III clinical trials are carried out without patient selection; therefore, it is hard to achieve personalized treatment and to monitor patient outcome individually. The trend for future clinical trials requires patient selection methods based on current understanding of GC biology with the application of biomarkers.
Collapse
Affiliation(s)
- Lian Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | | | | |
Collapse
|
147
|
Lee HE, Kim MA, Lee HS, Jung EJ, Yang HK, Lee BL, Bang YJ, Kim WH. MET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer 2012; 107:325-33. [PMID: 22644302 PMCID: PMC3394975 DOI: 10.1038/bjc.2012.237] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: The aim of this study was to compare gene copy number (GCN) and protein expression of MET and to evaluate their prognostic roles in gastric carcinomas. Methods: MET protein expression and gene amplification (GA) status were determined by immunohistochemistry (IHC) and silver in-situ hybridisation (SISH), respectively, in a large series of gastric carcinoma. Results: Protein overexpression was observed in 104 of 438 cases, with IHC 2+ in 94 and IHC 3+ in 10, and high polysomy of chromosome 7 and GA were found in 61 and 13 of 381, respectively. Direct comparison revealed a significant correlation between high level of protein expression and increased GCN. All cases with GA showed protein overexpression. Furthermore, all with IHC 3+ showed GA except 1, even which could be categorised as GA according to the ASCO/CAP guideline for human epidermal growth factor receptor 2 assessment. IHC 3+ and GA were significantly associated with poor prognosis. Conclusion: MET IHC reflects well on GA, and therefore, it could be a primary screening test for patient selection for anti-MET therapy if GA is a major determinant of drug responsiveness. Also, the prognostic role of MET indicates that anti-MET therapy is a very promising modality in adjuvant treatment for gastric cancer.
Collapse
Affiliation(s)
- H E Lee
- Department of Pathology, Seoul National University Hospital, 28 Yeongeon-dong, Jongno-gu, Seoul 110-799, Korea
| | | | | | | | | | | | | | | |
Collapse
|
148
|
|
149
|
|