201
|
Yhhu3813 is a novel selective inhibitor of c-Met kinase that inhibits c-Met-dependent neoplastic phenotypes of human cancer cells. Acta Pharmacol Sin 2014; 35:89-97. [PMID: 24241352 DOI: 10.1038/aps.2013.125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/02/2013] [Indexed: 01/17/2023] Open
Abstract
AIM c-Met kinase deregulation is strongly associated with the formation, progression and dissemination of human cancers. In this study we identified Yhhu3813 as a small-molecule inhibitor of c-Met kinase and characterized its antitumor properties both in vitro and in vivo. METHODS The activities of different kinases were measured using ELISA assays and signaling proteins in the cells were detected with Western blotting. Cell proliferation was assessed using SRB or MTT assay in twenty human cell lines and cell cycle distribution was determined with flow cytometry. Transwell-based assay was used to evaluate cell migration and invasion. Cell invasive growth was detected by a morphogenesis assay. c-Met overactivated human NSCLC cell line EBC-1 xenografts were used to evaluate the in vivo anti-tumor efficacy. RESULTS Yhhu3813 potently inhibited c-Met kinase activity in vitro with an IC50 value of 2.4±0.3 nmol/L, >400-fold higher than that for a panel of 15 different tyrosine kinases, suggesting a high selectivity of Yhhu3813. The compound (20, 100 and 500 nmol/L) dose-dependently inhibited the phosphorylation of c-Met and its key downstream Akt and Erk signal cascades in multiple c-Met aberrant human cancer cell lines, regardless of the mechanistic complexity in c-Met activation across different cellular contexts. In 20 human cancer cell lines harboring different backgrounds of c-Met expression/activation, Yhhu3813 potently inhibited c-Met-driven cell proliferation via arresting cells at G1/S phase. Furthermore, Yhhu3813 substantially impaired c-Met-mediated cell migration, invasion, scattering, and invasive growth. Oral administration of EBC-1 xenograft mice with Yhhu3813 (50 or 100 mg·kg(-1)·d(-1), qd, for 2 weeks) dose-dependently suppressed the tumor growth, which was correlated with a reduction in the intratumoral proliferation index and c-Met signaling. CONCLUSION Yhhu3813 is a potent selective inhibitor of c-Met that inhibits c-Met-dependent neoplastic phenotypes of human cancer cells in vitro and in vivo.
Collapse
|
202
|
Burtness B, Ilson D, Iqbal S. New directions in perioperative management of locally advanced esophagogastric cancer. Am Soc Clin Oncol Educ Book 2014:e172-e178. [PMID: 24857100 DOI: 10.14694/edbook_am.2014.34.e172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Cancers of the esophagus arise as adenocarcinomas and squamous cell carcinomas; these represent distinct diseases, with differing prognosis, yet they are often studied in common trials. With surgery alone, 5 year survival for T2-T3N0 disease is less than 30% to 40%, and declines to less than 25% with nodal involvement. The CROSS randomly assigned patients to surgery alone or to weekly carboplatin/paclitaxel X 5 and 41.4 Gy concurrent radiotherapy, followed by surgery. Seventy-five percent of enrolled patients had adenocarcinoma. Preoperative combined-modality therapy improved R0 resection from 69% to 92% (p < 0.001 and improved median survival from 24 months to 49.4 months (p < 0.003). This regimen reduced both locoregional recurrence (34% to 14%; p < 0.001) and the development of peritoneal carcinomatosis (14% to 4%; p < 0.001). Systemic perioperative therapy may have a greater effect on distant disease, the predominant mode of failure for these patients, and current trials compare preoperative chemoradiation with periooperative systemic therapy. PET scan response during preoperative chemotherapy without radiotherapy correlates with improvements in pathologic response and with improved survival. Nonresponse on early PET scan allows identifıcation of patients for earlier surgery and discontinuation of ineffective preoperative chemotherapy, without survival detriment. There is no predictive benefıt for early PET scan during the course of chemotherapy followed by chemoradiotherapy. The use of early PET scan during induction chemotherapy is being evaluated in CALGB/Alliance trial (NCT01333033). Molecular profıling has identifıed somatic gene mutations and pathways that may be oncogenic in upper gastrointestinal cancers. Potential targets include the epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), HER2, mammalian target of rapamycin (mTOR), fıbroblast growth factor receptor (FGFR), MEK, and others. Targeted therapies with known survival benefit in esophagogastric cancer are currently limited to trastuzumab for HER2 overexpressing cancers, or ramicirumab.
Collapse
Affiliation(s)
- Barbara Burtness
- From the Fox Chase Cancer Center, Philadelphia, PA; Memorial Sloan Kettering Cancer Center, New York, NY; University of Southern California, Los Angeles, CA
| | - David Ilson
- From the Fox Chase Cancer Center, Philadelphia, PA; Memorial Sloan Kettering Cancer Center, New York, NY; University of Southern California, Los Angeles, CA
| | - Syma Iqbal
- From the Fox Chase Cancer Center, Philadelphia, PA; Memorial Sloan Kettering Cancer Center, New York, NY; University of Southern California, Los Angeles, CA
| |
Collapse
|
203
|
Effects of MACC1 siRNA on biological behaviors of HeLa. Arch Gynecol Obstet 2013; 289:1271-80. [PMID: 24346124 DOI: 10.1007/s00404-013-3126-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 12/05/2013] [Indexed: 11/27/2022]
Abstract
PURPOSE Metastasis-associated in colon cancer 1 (MACC1) has been shown to play a critical role in several types of cancer. The purposes of this study were to evaluate MACC1 expression in cervical cancer and determine role of MACC1 small interference RNA (siRNA) in the growth and progression of cervical cancer. METHODS Immunohistochemical analysis of MACC1 expression was performed in different cervical lesion tissues. siRNA knockdown of MACC1 was performed. Cytoskeletal staining, RT-PCR, Western blot technology, transwell migration, MTT, and flow cytometry were used for identification of the functional roles of MACC1 siRNA in HeLa cells. RESULTS Immunohistochemistry demonstrated that MACC1 overexpression was detected in cervical cancer tissues. MACC1 siRNA transfection remarkably affected HeLa cell biological behaviors. Expression of MACC1 in HeLa cells was significantly down-regulated by MACC1 siRNA. In addition, knockdown of MACC1 in HeLa cells caused a significant decrease in cell proliferation or migration, and increased cell apoptosis rate. Flow cytometry showed that MACC1 siRNA may inhibit cell proliferation by interfering with cell mitosis and cell cycle progression. CONCLUSIONS These results suggest that MACC1 is a novel biomarker for cervical cancer diagnosis and a target for therapeutic interventions. Decreasing MACC1 expression by siRNA may prove to be an effective genetic therapy strategy.
Collapse
|
204
|
Abstract
Endocytosis entails selective packaging of cell-surface proteins, such as receptors for cytokines and adhesion components, in cytoplasmic vesicles (endosomes). The series of sorting events that determines the fate of internalized proteins, either degradation in lysosomes or recycling back to the plasma membrane, relies on intrinsic sequence motifs, posttranslational modifications (e.g., phosphorylation and ubiquitination), and transient assemblies of both Rab GTPases and phosphoinositide-binding proteins. This multicomponent process is enhanced and skewed in cancer cells; we review mechanisms enabling both major drivers of cancer, p53 and Ras, to bias recycling of integrins and receptor tyrosine kinases (RTKs). Likewise, cadherins and other junctional proteins of cancer cells are constantly removed from the cell surface, thereby disrupting tissue polarity and instigating motile phenotypes. Mutant forms of RTKs able to evade Cbl-mediated ubiquitination, along with overexpression of the wild-type forms and a variety of defective feedback regulatory loops, are frequently detected in tumors. Finally, we describe pharmacological attempts to harness the peculiar endocytic system of cancer, in favor of effective patient treatment.
Collapse
|
205
|
Sundaram S, Freemerman AJ, Johnson AR, Milner JJ, McNaughton KK, Galanko JA, Bendt KM, Darr DB, Perou CM, Troester MA, Makowski L. Role of HGF in obesity-associated tumorigenesis: C3(1)-TAg mice as a model for human basal-like breast cancer. Breast Cancer Res Treat 2013; 142:489-503. [PMID: 24218051 DOI: 10.1007/s10549-013-2741-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 10/16/2013] [Indexed: 11/29/2022]
Abstract
Obesity is associated with basal-like breast cancer (BBC), an aggressive breast cancer subtype. The objective of this study was to determine whether obesity promotes BBC onset in adulthood and to evaluate the role of stromal-epithelial interactions in obesity-associated tumorigenesis. We hypothesized that hepatocyte growth factor (HGF) plays a promoting role in BBC, which express the HGF receptor, c-Met. In C3(1)-T(Ag) mice, a murine model of BBC, we demonstrated that obesity leads to a significant increase in HGF secretion and an associated decrease in tumor latency. By immunohistochemical analysis, normal mammary gland exhibited obesity-induced HGF, c-Met and phospho-c-Met, indicating that the activation of the cascade was obesity-driven. HGF secretion was also increased from primary mammary fibroblasts isolated from normal mammary glands and tumors of obese mice compared to lean. These results demonstrate that obesity-induced elevation of HGF expression is a stable phenotype, maintained after several passages, and after removal of dietary stimulation. Conditioned media from primary tumor fibroblasts from obese mice drove tumor cell proliferation. In co-culture, neutralization of secreted HGF blunted tumor cell migration, further linking obesity-mediated HGF-dependent effects to in vitro measures of tumor aggressiveness. In sum, these results demonstrate that HGF/c-Met plays an important role in obesity-associated carcinogenesis. Understanding the effects of obesity on risk and progression is important given that epidemiologic studies imply a portion of BBC could be eliminated by reducing obesity.
Collapse
Affiliation(s)
- Sneha Sundaram
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, CB 7461, 2203 McGavran Greenberg Hall, Chapel Hill, NC, 27599-7461, USA,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Jin Y, Sun PL, Kim H, Seo AN, Jheon S, Lee CT, Chung JH. MET Gene Copy Number Gain is an Independent Poor Prognostic Marker in Korean Stage I Lung Adenocarcinomas. Ann Surg Oncol 2013; 21:621-8. [DOI: 10.1245/s10434-013-3355-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Indexed: 12/11/2022]
|
207
|
de Mello RA, Marques AM, Araújo A. HER2 therapies and gastric cancer: a step forward. World J Gastroenterol 2013; 19:6165-9. [PMID: 24115812 PMCID: PMC3787345 DOI: 10.3748/wjg.v19.i37.6165] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/25/2013] [Accepted: 08/12/2013] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer usually is diagnosed in advanced stages and thus current medical practice affords limited therapeutic options. However, recent studies established the role of human epidermal growth factor receptor 2 (HER2) in clinical management. Trastuzumab, an anti-HER2 monoclonal antibody, acquired a main role in advanced gastric cancer harboring HER2 overexpression and/or amplification improving survival to 17.1 mo according to trastuzumab for gastric cancer phase III trial results. Also, new promising drugs, such as c-Met inhibitors, are in development and assessment for this setting. Certainly, novel drugs will emerge in the next feel years for help oncologists improve clinical management of advanced gastric cancer providing higher survival and quality of life. In this mini-review we will discuss some issues in this regard and provide an actual overview of this setting.
Collapse
|
208
|
Bossi P, Locati L, Licitra L. Emerging tyrosine kinase inhibitors for head and neck cancer. Expert Opin Emerg Drugs 2013; 18:445-59. [DOI: 10.1517/14728214.2013.842976] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
209
|
Jacobson O, Chen X. Interrogating tumor metabolism and tumor microenvironments using molecular positron emission tomography imaging. Theranostic approaches to improve therapeutics. Pharmacol Rev 2013; 65:1214-56. [PMID: 24064460 DOI: 10.1124/pr.113.007625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Positron emission tomography (PET) is a noninvasive molecular imaging technology that is becoming increasingly important for the measurement of physiologic, biochemical, and pharmacological functions at cellular and molecular levels in patients with cancer. Formation, development, and aggressiveness of tumor involve a number of molecular pathways, including intrinsic tumor cell mutations and extrinsic interaction between tumor cells and the microenvironment. Currently, evaluation of these processes is mainly through biopsy, which is invasive and limited to the site of biopsy. Ongoing research on specific target molecules of the tumor and its microenvironment for PET imaging is showing great potential. To date, the use of PET for diagnosing local recurrence and metastatic sites of various cancers and evaluation of treatment response is mainly based on [(18)F]fluorodeoxyglucose ([(18)F]FDG), which measures glucose metabolism. However, [(18)F]FDG is not a target-specific PET tracer and does not give enough insight into tumor biology and/or its vulnerability to potential treatments. Hence, there is an increasing need for the development of selective biologic radiotracers that will yield specific biochemical information and allow for noninvasive molecular imaging. The possibility of cancer-associated targets for imaging will provide the opportunity to use PET for diagnosis and therapy response monitoring (theranostics) and thus personalized medicine. This article will focus on the review of non-[(18)F]FDG PET tracers for specific tumor biology processes and their preclinical and clinical applications.
Collapse
Affiliation(s)
- Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD.
| | | |
Collapse
|
210
|
Msaouel P, Nandikolla G, Pneumaticos SG, Koutsilieris M. Bone microenvironment-targeted manipulations for the treatment of osteoblastic metastasis in castration-resistant prostate cancer. Expert Opin Investig Drugs 2013; 22:1385-400. [PMID: 24024652 DOI: 10.1517/13543784.2013.824422] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Most patients with advanced prostate cancer will develop incurable bone metastasis. Although prostate cancer is the quintessential androgen-dependent neoplastic disease in males, the tumor will ultimately become refractory to androgen ablation treatment. Understanding the complex dialog between prostate cancer and the bone microenvironment has allowed the development of promising treatment strategies. AREAS COVERED The present review summarizes the pathophysiology of prostate cancer bone metastasis and provides a concise update on bone microenvironment-targeted therapies for prostate cancer. The current and future prospects and challenges of these strategies are also discussed. EXPERT OPINION A wide variety of signaling pathways, bone turnover homeostatic mechanisms and immunoregulatory networks are potential targets for the treatment of metastatic castration-resistant prostate cancer (mCRPC). Anti-survival factor therapy can enhance the efficacy of existing treatment regimens for mCRPC by exploiting the interaction between the bone microenvironment and androgen signaling networks. In addition, many novel bone microenvironment-targeted strategies have produced promising objective clinical responses. Further elucidation of the complex interactions between prostate cancer cells and the bone stroma will open up new avenues for treatment interventions that can produce sustained cancer suppression.
Collapse
Affiliation(s)
- Pavlos Msaouel
- Jacobi Medical Center, Department of Internal Medicine, Albert Einstein College of Medicine , Bronx, NY , USA
| | | | | | | |
Collapse
|
211
|
Pérez-Vargas JCS, Biondani P, Maggi C, Gariboldi M, Gloghini A, Inno A, Volpi CC, Gualeni AV, di Bartolomeo M, de Braud F, Castano A, Bossi I, Pietrantonio F. Role of cMET in the development and progression of colorectal cancer. Int J Mol Sci 2013; 14:18056-77. [PMID: 24005867 PMCID: PMC3794769 DOI: 10.3390/ijms140918056] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 08/13/2013] [Accepted: 08/27/2013] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal-epithelial transition (MET) is a member of a distinct subfamily of heterodimeric receptor tyrosine kinase receptors that specifically binds the hepatocyte growth factor (HGF). Binding to HGF leads to receptor dimerization/multimerization and phosphorylation, resulting in its catalytic activation. MET activation drives the malignant progression of several tumor types, including colorectal cancer (CRC), by promoting signaling cascades that mainly result in alterations of cell motility, survival, and proliferation. MET is aberrantly activated in many human cancers through various mechanisms, including point mutations, gene amplification, transcriptional up-regulation, or ligand autocrine loops. MET promotes cell scattering, invasion, and protection from apoptosis, thereby acting as an adjuvant pro-metastatic gene for many tumor types. In CRC, MET expression confers more aggressiveness and worse clinical prognosis. With all of this rationale, inhibitors that target the HGF/MET axis with different types of response have been developed. HGF and MET are new promising targets to understand the pathogenesis of CRC and for the development of new, targeted therapies.
Collapse
Affiliation(s)
| | - Pamela Biondani
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mails: (P.B.); (C.M.); (M.B.); (F.B.); (A.C.); (I.B.)
| | - Claudia Maggi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mails: (P.B.); (C.M.); (M.B.); (F.B.); (A.C.); (I.B.)
| | - Manuela Gariboldi
- Experimental Oncology and Molecular Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mail:
- FIRC Institute of Molecolar Oncology Foundation (IFOM), 1-20133 Milan, Italy
| | - Annunziata Gloghini
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mails: (A.G.); (C.C.V.); (A.V.G.)
| | - Alessandro Inno
- Medical Oncology, Sacro Cuore-Don Calabria Hospital, 37024 Negrar (Verona), Italy; E-Mail:
| | - Chiara Costanza Volpi
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mails: (A.G.); (C.C.V.); (A.V.G.)
| | - Ambra Vittoria Gualeni
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mails: (A.G.); (C.C.V.); (A.V.G.)
| | - Maria di Bartolomeo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mails: (P.B.); (C.M.); (M.B.); (F.B.); (A.C.); (I.B.)
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mails: (P.B.); (C.M.); (M.B.); (F.B.); (A.C.); (I.B.)
| | - Alessandra Castano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mails: (P.B.); (C.M.); (M.B.); (F.B.); (A.C.); (I.B.)
| | - Ilaria Bossi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mails: (P.B.); (C.M.); (M.B.); (F.B.); (A.C.); (I.B.)
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1-20133 Milan, Italy; E-Mails: (P.B.); (C.M.); (M.B.); (F.B.); (A.C.); (I.B.)
| |
Collapse
|
212
|
|
213
|
Minuti G, D'Incecco A, Cappuzzo F. Targeted therapy for NSCLC with driver mutations. Expert Opin Biol Ther 2013; 13:1401-12. [PMID: 23930754 DOI: 10.1517/14712598.2013.827657] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Activating mutations of the epidermal growth factor receptor (EGFR) gene and rearrangement of anaplastic lymphoma kinase (ALK) gene best illustrate the therapeutic relevance of molecular characterization in non-small cell lung cancer (NSCLC) patients. Several genetic aberrations with a potential prognostic or predictive role have been identified, mainly in adenocarcinoma subtype, including ROS1, RET, MET, HER2, BRAF and KRAS. More recently oncogenic drivers, such as DDR2, FGFR1 and PI3KCA, have been characterized in squamous cell lung carcinoma (SCC) and target agents are currently under evaluation. The aim of this review is to summarize the growing scenario of new targetable oncogenes in NSCLC. AREAS COVERED For this review article all published data on NSCLC genomic alterations, including the techniques employed for oncogenic drivers identification, the prevalence of each one in lung cancer subtypes, the preclinical data corroborating their role in tumorigenesis and the potential biological tailored agents tested and under evaluation were collected and analyzed using PubMed. EXPERT OPINION Oncogenic products represent reliable targets for drug therapy and the expanding knowledge of molecular pathways involved in lung tumorigenesis is resulting in a dramatic change of treatment strategies leading to an improvement in disease and symptom control, extending life duration and improving quality of life.
Collapse
Affiliation(s)
- Gabriele Minuti
- Medical Oncology Department, Civil Hospital of Livorno, Istituto Toscano Tumori , Viale Alfieri 36, 57100, Livorno , Italy +39 0586 223189 ; +39 0586 223457 ;
| | | | | |
Collapse
|
214
|
Abstract
PURPOSE OF REVIEW The mesenchymal-epidermal transition (c-MET) receptor tyrosine kinase has a central role in the cancer cell's survival. MET and its ligand, hepatocyte growth factor (HGF), have recently been identified as promising targets in solid tumors, including nonsmall-cell lung cancer (NSCLC). RECENT FINDINGS Aberrant MET activation can be the result of different mechanisms such as MET and HGF overexpression, MET gene amplification or mutation. Retrospective studies in NSCLC showed that MET gene copy number is a negative prognostic factor, although few data are available on the role of MET mutations. In preclinical models, cell lines with MET gene amplification are extremely sensitive to MET inhibition. Although the inner presence of gene amplification is a rare event (1-7% cases), MET amplification has emerged as one of the critical events for acquired resistance in epidermal growth factor receptor (EGFR) mutated lung adenocarcinomas refractory to EGFR-tyrosine kinase inhibitors (TKIs). In NSCLC with acquired resistance to EGFR-TKIs, MET amplification occurs in up to 20% cases and preclinical and clinical data indicated MET and EGFR co-inhibition as a potential effective strategy to overcome resistance. SUMMARY MET has recently emerged as a promising target, and ongoing trials will clarify the role of anti-MET strategies in NSCLC.
Collapse
|
215
|
Yan B, Lim M, Zhou L, Kuick CH, Leong MY, Yong KJ, Aung L, Salto-Tellez M, Chang KTE. Identification of MET genomic amplification, protein expression and alternative splice isoforms in neuroblastomas. J Clin Pathol 2013; 66:985-91. [PMID: 23801497 DOI: 10.1136/jclinpath-2012-201375] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Crizotinib, a dual anaplastic lymphoma kinase (ALK) and mesenchymal-epithelial transition (MET) tyrosine kinase inhibitor, is currently being evaluated for the treatment of neuroblastoma. Its effects are thought to be mediated mainly via its activity against ALK. Although MET genomic/protein expression status might conceivably affect crizotinib efficacy, this issue has hitherto not received attention in neuroblastomas. AIMS/METHODS MET genomic and protein expression status was characterised by silver in situ hybridisation and immunohistochemistry (IHC) respectively, in a cohort of 54 neuroblastoma samples. MET splice isoforms were characterised in 15 of these samples by quantitative PCR. RESULTS One case (1/54; prevalence 1.85%) displayed MET genomic amplification, while another case (1/54; prevalence 1.85%) displayed strong membranous MET protein expression (IHC score 3+). Alternative exon 10-deleted and exon 14-deleted MET splice isoforms were identified. CONCLUSIONS MET amplification and protein expression, although low in prevalence, are present in neuroblastomas. This has implications when crizotinib is employed as a therapeutic agent in neuroblastomas. Additionally, the existence of alternatively spliced MET isoforms may have clinical and biological implications in neuroblastomas.
Collapse
Affiliation(s)
- Benedict Yan
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Landi L, Minuti G, D’Incecco A, Salvini J, Cappuzzo F. MET overexpression and gene amplification in NSCLC: a clinical perspective. LUNG CANCER (AUCKLAND, N.Z.) 2013; 4:15-25. [PMID: 28210131 PMCID: PMC5217438 DOI: 10.2147/lctt.s35168] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transmembrane tyrosine kinase mesenchymal-epidermal transition (MET) receptor and its ligand, hepatocyte growth factor, also known as scatter factor, have recently been identified as novel promising targets in several human malignancies, including non-small cell lung cancer (NSCLC). Amplification, mutation, or overexpression of the MET gene can result in aberrant activation of the MET axis, leading to migration, invasion, proliferation, metastasis, and neoangiogenesis of cancer cells, suggesting that interfering with the MET/hepatocyte growth factor pathway could represent a potential antitumor strategy. While the role of MET mutations in NSCLC is not as yet fully understood, retrospective studies have shown that an increased MET gene copy number is a negative prognostic factor. In NSCLC, amplification of the MET gene is a relatively rare event, occurring in approximately 4% of patients not previously exposed to systemic therapies and in up to 20% of patients with acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors. In preclinical models, the presence of MET amplification is a predictor of high sensitivity to anti-MET compounds, and several agents have entered in clinical trials for patients having advanced disease, with promising results. The aim of the present review is to summarize available data on the role of MET in NSCLC and to describe therapeutic strategies under investigation.
Collapse
Affiliation(s)
- Lorenza Landi
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| | - Gabriele Minuti
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| | - Armida D’Incecco
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| | - Jessica Salvini
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| | - Federico Cappuzzo
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| |
Collapse
|
217
|
Ding X, Pan H, Li J, Zhong Q, Chen X, Dry SM, Wang CY. Epigenetic activation of AP1 promotes squamous cell carcinoma metastasis. Sci Signal 2013; 6:ra28.1-13, S0-15. [PMID: 23633675 DOI: 10.1126/scisignal.2003884] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The transcription factor AP1 (activating protein 1), a heterodimer of the JUN and FOS proteins, promotes the invasive growth and metastasis of various tumors such as squamous cell carcinoma (SCC), breast cancer, and melanoma. AP1 activity is transcriptionally induced through a positive feedback loop. We identified the histone demethylase KDM4A (lysine-specific demethylase 4A) as a key epigenetic priming factor in this positive feedback loop. KDM4A contributed to the induction of genes encoding the AP1 transcription factors and the invasive growth and metastasis of SCC. KDM4A knockdown decreased the growth factor-induced messenger RNA expression and protein abundance of AP1 family members, including JUN and FOSL1. Mechanistically, histone demethylation by KDM4A facilitated the binding of the AP1 complex to the promoters of JUN and FOSL1, thereby promoting the positive feedback loop that maintains activation of AP1. In a mouse model of SCC, KDM4A knockdown inhibited lymph node metastasis. Moreover, the abundance of KDM4A correlated with the abundance of JUN and FOSL1 in human SCC tissues, and KDM4A expression was increased in human lymph node metastases. Our studies provide insights into the epigenetic control of AP1 and tumor invasion and suggest that KDM4A could be an important therapeutic target for inhibiting invasive SCC growth and metastasis.
Collapse
Affiliation(s)
- Xiangming Ding
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | |
Collapse
|
218
|
Kristensen IB, Christensen JH, Lyng MB, Møller MB, Pedersen L, Rasmussen LM, Ditzel HJ, Abildgaard N. Hepatocyte growth factor pathway upregulation in the bone marrow microenvironment in multiple myeloma is associated with lytic bone disease. Br J Haematol 2013; 161:373-82. [PMID: 23431957 DOI: 10.1111/bjh.12270] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 01/21/2013] [Indexed: 12/17/2022]
Abstract
Lytic bone disease (LBD) in multiple myeloma (MM) is caused by osteoclast hyperactivation and osteoblast inhibition. Based on in vitro studies, the hepatocyte growth factor (HGF) pathway is thought to be central in osteoblast inhibition. We evaluated the gene expression of the HGF pathway in vivo using bone marrow biopsies (BMBs) of patients with MM and monoclonal gammopathy of undetermined significance (MGUS), and healthy volunteers (HV). BMBs (N = 110) obtained at diagnosis were snap-frozen and used to evaluate gene expression by quantitative reverse transcription polymerase chain reaction. LBD was evaluated using standard radiographs. Enzyme-linked immunosorbent assay (ELISA) was performed on matched bone marrow plasma and immunohistochemistry on matched formalin-fixed paraffin-embedded biopsies. Gene expression of HGF, SDC1, and MET in BMBs were significantly altered in MM versus HV and MGUS, and HGF and MET correlated with the extent of LBD. A significant correlation between gene and protein expression levels was observed for SDC1 (Syndecan-1) and HGF. The HGF bone marrow plasma level was significantly lower in MM patients with no/limited versus advanced LBD. Our novel approach using snap-frozen BMBs seems generally applicable because it allows evaluation of gene expression independent of the extent of MM plasma-cell infiltration. Our study highlights the importance of the HGF pathway in MM LBD.
Collapse
Affiliation(s)
- Ida B Kristensen
- Department of Haematology, Odense University Hospital, Odense, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
219
|
Goodin S, DiPaola RS. Cabozantinib in Prostate Cancer: The Beginning of a Precision Paradigm? J Clin Oncol 2013; 31:401-3. [DOI: 10.1200/jco.2012.46.6185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Susan Goodin
- The Cancer Institute of New Jersey; and University of Medicine and Dentistry of New Jersey Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Robert S. DiPaola
- The Cancer Institute of New Jersey; and University of Medicine and Dentistry of New Jersey Robert Wood Johnson Medical School, New Brunswick, NJ
| |
Collapse
|
220
|
Abstract
Under normal physiological conditions, the hepatocyte growth factor (HGF) and its receptor, the MET transmembrane tyrosine kinase (cMET), are involved in embryogenesis, morphogenesis, and wound healing. The HGF-cMET axis promotes cell survival, proliferation, migration, and invasion via modulation of epithelial-mesenchymal interactions. Hepatocellular cancer (HCC) is the third most common cause of worldwide cancer-related mortality; advanced disease is associated with a paucity of therapeutic options and a five-year survival rate of only 10%. Dysregulation of the HGF-cMET pathway is implicated in HCC carcinogenesis and progression through activation of multiple signaling pathways; therefore, cMET inhibition is a promising therapeutic strategy for HCC treatment. The authors review HGF-cMET structure and function in normal tissue and in HCC, cMET inhibition in HCC, and future strategies for biomarker identification.
Collapse
|
221
|
Shieh JM, Tang YA, Yang TH, Chen CY, Hsu HS, Tan YHC, Salgia R, Wang YC. Lack of association of C-Met-N375S sequence variant with lung cancer susceptibility and prognosis. Int J Med Sci 2013; 10:988-94. [PMID: 23801885 PMCID: PMC3691797 DOI: 10.7150/ijms.5944] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 05/29/2013] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Previously, we identified a sequence variant (N375S) of c-Met gene, however, its association with lung cancer risk and prognosis remain undefined. PATIENTS AND METHODS We investigated the genotype distribution of the c-Met-N375S sequence variant in 206 lung cancer patients and 207 non-cancer controls in the Taiwanese population by DNA sequencing. RESULTS Lung cancer patients with variant A/G and G/G genotypes showed 1.08-fold increased cancer risk when compared to patients with the wild-type A/A genotype (95% CI, 0.60-1.91). There were no significant differences in postoperative survival between c-Met-N375S and wild-type patients. In the cell model, the c-Met-N375S cells showed a decrease in cell death upon treatment with MET inhibitor SU11274 compared to wild-type cells. CONCLUSION Our data suggest that the c-Met-N375S sequence variant may not play a significant role in cancer susceptibility and the prognosis of lung cancer patients. The correlation with chemoresponse of c-Met-N375S is worth further investigation in patients receiving MET therapy.
Collapse
Affiliation(s)
- Jiunn-Min Shieh
- 1. Department of Chest Medicine, Chi Mei Medical Center, Tainan, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
222
|
Schöpper S, Laban S, Güldenzoph B, Münscher A, Knecht R, Kurzweg T. [New aspects in the therapy of recurrence and metastasis of head and neck cancer: highlights from the 2012 ASCO meeting]. HNO 2012; 60:957-61. [PMID: 23114541 DOI: 10.1007/s00106-012-2596-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The majority of patients with a squamous cell carcinoma of the head and neck region (HNSCC) initially present in a locally advanced stage of the disease. Despite aggressive protocols in first-line treatment, some of these patients develop locoregional recurrences or metastases and are, in particular, extraordinarily challenging for the multidisciplinary treatment team. Therefore, among resectability and prior therapy, age, performance status and individual expectations of the patient have to be taken into account. Apart from surgical options like salvage surgery, chemotherapy and target therapy as well as reirradiation are possible treatment concepts. Unfortunately, most treatment options offer only little to no survival benefit.
Collapse
Affiliation(s)
- S Schöpper
- Klinik und Poliklinik für Hals-, Nasen- und Ohrenheilkunde, Kopf-Hals-Tumorzentrum des Universitären Cancer Center Hamburg, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Deutschland.
| | | | | | | | | | | |
Collapse
|