151
|
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: 154] [Impact Index Per Article: 12.8] [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
|
152
|
Zhang Y, Farenholtz KE, Yang Y, Guessous F, Dipierro CG, Calvert VS, Deng J, Schiff D, Xin W, Lee JK, Purow B, Christensen J, Petricoin E, Abounader R. Hepatocyte growth factor sensitizes brain tumors to c-MET kinase inhibition. Clin Cancer Res 2013; 19:1433-44. [PMID: 23386689 DOI: 10.1158/1078-0432.ccr-12-2832] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE The receptor tyrosine kinase (RTK) c-MET and its ligand hepatocyte growth factor (HGF) are deregulated and promote malignancy in cancer and brain tumors. Consequently, clinically applicable c-MET inhibitors have been developed. The purpose of this study was to investigate the not-well-known molecular determinants that predict responsiveness to c-MET inhibitors and to explore new strategies for improving inhibitor efficacy in brain tumors. EXPERIMENTAL DESIGN We investigated the molecular factors and pathway activation signatures that determine sensitivity to c-MET inhibitors in a panel of glioblastoma and medulloblastoma cells, glioblastoma stem cells, and established cell line-derived xenografts using functional assays, reverse protein microarrays, and in vivo tumor volume measurements, but validation with animal survival analyses remains to be done. We also explored new approaches for improving the efficacy of the inhibitors in vitro and in vivo. RESULTS We found that HGF coexpression is a key predictor of response to c-MET inhibition among the examined factors and identified an ERK/JAK/p53 pathway activation signature that differentiates c-MET inhibition in responsive and nonresponsive cells. Surprisingly, we also found that short pretreatment of cells and tumors with exogenous HGF moderately but statistically significantly enhanced the antitumor effects of c-MET inhibition. We observed a similar ligand-induced sensitization effect to an EGF receptor small-molecule kinase inhibitor. CONCLUSIONS These findings allow the identification of a subset of patients that will be responsive to c-MET inhibition and propose ligand pretreatment as a potential new strategy for improving the anticancer efficacy of RTK inhibitors.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia 22908, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Wu D, Yu H, Li J. [Mechanisms of resistance to EML4-ALK inhibitors in non-small cell lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2013; 16:48-53. [PMID: 23327874 PMCID: PMC6000462 DOI: 10.3779/j.issn.1009-3419.2013.01.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Di Wu
- Department of Medical Oncology, Jilin Province Tumor Hospital, Changchun 130012, China;Tumor Center, First Hospital of Jilin University, Changchun 130021, China
| | | | | |
Collapse
|
154
|
Lackner MR, Wilson TR, Settleman J. Mechanisms of acquired resistance to targeted cancer therapies. Future Oncol 2012; 8:999-1014. [PMID: 22894672 DOI: 10.2217/fon.12.86] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Drugs that target genomically defined vulnerabilities in human tumors have now been clinically validated as effective cancer therapies. However, the relatively rapid acquisition of resistance to such treatments that is observed in virtually all cases significantly limits their utility and remains a substantial challenge to the clinical management of advanced cancers. As molecular mechanisms of resistance have begun to be elucidated, new strategies to overcome or prevent the development of resistance have begun to emerge. In some cases, specific mutational mechanisms contribute directly to acquired drug resistance, and in other cases it appears that nonmutational and possibly epigenetic mechanisms play a significant role. This article discusses the various genetic and nongenetic mechanisms of acquired drug resistance that have been reported in the context of 'rationally targeted' drug therapies.
Collapse
Affiliation(s)
- Mark R Lackner
- Department of Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | | | | |
Collapse
|
155
|
Ach T, Zeitler K, Schwarz-Furlan S, Baader K, Agaimy A, Rohrmeier C, Zenk J, Gosau M, Reichert TE, Brockhoff G, Ettl T. Aberrations of MET are associated with copy number gain of EGFR and loss of PTEN and predict poor outcome in patients with salivary gland cancer. Virchows Arch 2012; 462:65-72. [PMID: 23242174 DOI: 10.1007/s00428-012-1358-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 11/12/2012] [Accepted: 12/03/2012] [Indexed: 12/28/2022]
Abstract
Hepatocyte growth factor receptor (MET) is a key driver of oncogenic transformation. Copy number gain and amplification of MET positively enhance tumour growth, invasiveness and metastasis in different cancer types. In the present study, 266 carcinomas of the major and minor salivary glands were investigated for genomic MET status by fluorescence in situ hybridization and for protein expression by immunohistochemistry. Results were matched with clinicopathological parameters, long-term survival and the status of epidermal growth factor receptor (EGFR) and phosphatase and tensin homologue (PTEN). Low polysomy (n = 42), high polysomy (n = 27), amplification (n = 2) and deletion (n = 18) were found as aberrations of genomic MET in certain subtypes. MET aberrations were associated with increased patient age (>70 years, p = 0.003), male gender (p = 0.01), increased tumour size (p = 0.002), lymph node metastases (p < 0.001), high-grade malignancy (p < 0.001) and unfavourable overall survival (p < 0.001). Both copy number gain (p < 0.001) and deletion (p = 0.031) of MET correlated with copy number gain of EGFR. Tumours with genomic loss of PTEN (n = 48) concurrently presented aberration of genomic MET (p < 0.001). MET gene status significantly correlated with protein status (p = 0.038). In conclusion, gain but also loss of genomic MET activity correlates with aggressive tumour growth, nodal metastasis and worse overall survival in salivary gland cancer. Moreover, aberrations of MET are associated with EGFR and PTEN signalling and might possess relevance for targeted therapies of salivary gland carcinomas in the future.
Collapse
Affiliation(s)
- Tobias Ach
- Department of Oral and Maxillofacial Surgery, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Development of PI3K/AKT/mTOR pathway inhibitors and their application in personalized therapy for non-small-cell lung cancer. J Thorac Oncol 2012; 7:1315-26. [PMID: 22648207 DOI: 10.1097/jto.0b013e31825493eb] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lung cancer is a common disease with more than 1.6 million new cases diagnosed worldwide in 2008. Treatments for patients with advanced disease are rarely curative, and responses to therapy are often followed by relapse, which highlights the large unmet need for novel therapies. Recent advances in cancer treatment have focused on personalized therapy, whereby patients are treated with agents that best target the molecular drivers of their disease. Thus, a better understanding of the pathways that drive cancer or drug resistance is of critical importance. One such example is the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, which is activated in many lung cancer patients and represents a target for therapy. PI3K/AKT/mTOR pathway activation has also been observed in tumors resistant to agents targeting upstream receptor tyrosine kinases. Agents that target this pathway have the potential to shut down survival pathways, and are being explored both in the setting of pathway-activating mutations and for their ability to restore sensitivity to upstream signaling targeted agents. Here, we examine the frequency of mutations activating the PI3K/AKT/mTOR pathway, review the novel agents being explored to target this pathway, and explore the potential role of the inhibition of this pathway in the clinical development of these agents.
Collapse
|
157
|
Bagrodia S, Smeal T, Abraham RT. Mechanisms of intrinsic and acquired resistance to kinase-targeted therapies. Pigment Cell Melanoma Res 2012; 25:819-31. [PMID: 22883054 DOI: 10.1111/pcmr.12007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Revised: 08/07/2012] [Indexed: 12/19/2022]
Abstract
Cancer drugs that target pivotal signaling molecules required for malignant cell survival and growth have demonstrated striking antitumor activities in appropriately selected patient populations. Unfortunately, however, therapeutic responses are often of limited duration, typically 6-12 months, because of emergence of drug-resistant subclones of tumor cells. In this review, we highlight several of the mechanisms of emergent resistance to several kinase-targeted small molecule therapies used in melanoma, non-small cell lung cancer (NSCLC) and other solid tumors as illustrative examples. We discuss the implications of these findings for the development of new treatment strategies to delay or prevent the onset of drug resistance.
Collapse
Affiliation(s)
- Shubha Bagrodia
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10777 Science Center Drive, San Diego, CA, USA
| | | | | |
Collapse
|
158
|
Abstract
The receptor tyrosine kinase c-MET and its ligand, hepatocyte growth factor (HGF), regulate multiple cellular processes that stimulate cell proliferation, invasion and angiogenesis. This review provides an overview of the evidence to support c-MET or the HGF/c-MET signaling pathway as relevant targets for personalized cancer treatment based on high frequencies of c-MET and/or HGF overexpression, activation, amplification in non-small cell lung carcinoma (NSCLC), gastric, ovarian, pancreatic, thyroid, breast, head and neck, colon and kidney carcinomas. Additionally, the current knowledge of small molecule inhibitors (tivantinib [ARQ 197]), c-MET/HGF antibodies (rilotumumab and MetMAb) and mechanisms of resistance to c-MET-targeted therapies are discussed.
Collapse
Affiliation(s)
- J Rafael Sierra
- Princess Margaret Hospital/Ontario Cancer Institute and University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
159
|
Belalcazar A, Azaña D, Perez CA, Raez LE, Santos ES. Targeting the Met pathway in lung cancer. Expert Rev Anticancer Ther 2012; 12:519-28. [PMID: 22500688 DOI: 10.1586/era.12.16] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Dysregulation of Met signaling has been implicated in the initiation, progression and metastasis of human cancers, and therefore represents an attractive target for anticancer drug development. Met is overexpressed in non-small-cell lung cancer and its lack of staining in normal lung tissue makes it an attractive target. To date, erlotinib and gefitinib have established themselves as first-line therapy for non-small-cell lung cancer patients whose tumors harbor an EGF receptor gene mutation, and hence, it is crucial that we identify mechanisms of resistance that could be targeted by novel agents, while keeping an acceptable toxicity profile at the same time; something very important when we develop these new drugs. Inhibitors of the Met pathway represent a therapeutic alternative in this setting. In this review, we discuss the early clinical studies reported using two Met inhibitors, a monoclonal antibody (MetMAb) and a small molecule tyrosine kinase inhibitor (MGCD265).
Collapse
Affiliation(s)
- Astrid Belalcazar
- Department of Internal Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | | | | |
Collapse
|
160
|
Locatelli A, Lofgren KA, Daniel AR, Castro NE, Lange CA. Mechanisms of HGF/Met signaling to Brk and Sam68 in breast cancer progression. Discov Oncol 2012; 3:14-25. [PMID: 22124844 DOI: 10.1007/s12672-011-0097-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Signal transduction pathways downstream of receptor tyrosine kinases (RTKs) are often deregulated during oncogenesis, tumor progression, and metastasis. In particular, the peptide growth factor hormone, hepatocyte growth factor (HGF), and its specific receptor, Met tyrosine kinase, regulate cancer cell migration, thereby conferring an aggressive phenotype (Nakamura et al., J Clin Invest 106(12):1511-1519, 2000; Huh et al., Proc Natl Acad Sci U S A 101:4477-4482, 2004). Additionally, overexpression of Met is associated with enhanced invasiveness of breast cancer cells (Edakuni et al., Pathol Int 51(3):172-178, 2001; Jin et al., Cancer 79(4):749-760, 1997; Tuck et al., Am J Pathol 148(1):225-232, 1996). Here, we review the regulation of recently identified novel downstream mediators of HGF/Met signaling, Breast tumor kinase (Brk/PTK6), and Src-associated substrate during mitosis of 68 kDa (Sam68), and discuss their relevance to mechanisms of breast cancer progression.
Collapse
Affiliation(s)
- Alessia Locatelli
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, 55455, USA
| | | | | | | | | |
Collapse
|
161
|
Harbinski F, Craig VJ, Sanghavi S, Jeffery D, Liu L, Sheppard KA, Wagner S, Stamm C, Buness A, Chatenay-Rivauday C, Yao Y, He F, Lu CX, Guagnano V, Metz T, Finan PM, Hofmann F, Sellers WR, Porter JA, Myer VE, Graus-Porta D, Wilson CJ, Buckler A, Tiedt R. Rescue Screens with Secreted Proteins Reveal Compensatory Potential of Receptor Tyrosine Kinases in Driving Cancer Growth. Cancer Discov 2012; 2:948-59. [DOI: 10.1158/2159-8290.cd-12-0237] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
162
|
Blumenschein GR, Mills GB, Gonzalez-Angulo AM. Targeting the hepatocyte growth factor-cMET axis in cancer therapy. J Clin Oncol 2012; 30:3287-96. [PMID: 22869872 DOI: 10.1200/jco.2011.40.3774] [Citation(s) in RCA: 224] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The hepatocyte growth factor (HGF) and its receptor, the transmembrane tyrosine kinase cMET, promote cell proliferation, survival, motility, and invasion as well as morphogenic changes that stimulate tissue repair and regeneration in normal cells but can be co-opted during tumor growth. MET overexpression, with or without gene amplification, has been reported in a variety of human cancers, including breast, lung, and GI malignancies. Furthermore, high levels of HGF and/or cMET correlate with poor prognosis in several tumor types, including breast, ovarian, cervical, gastric, head and neck, and non-small-cell lung cancers. Gene amplification and protein overexpression of cMET drive resistance to epidermal growth factor receptor family inhibitors, both in preclinical models and in patients. It is increasingly apparent that the HGF-cMET axis signaling network is complex, and rational combinatorial therapy is needed for optimal clinical efficacy. Better understanding of HGF-cMET axis signaling and the mechanism of action of HGF-cMET inhibitors, along with the identification of biomarkers of response and resistance, will lead to more effective targeting of this pathway for cancer therapy.
Collapse
Affiliation(s)
- George R Blumenschein
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Box 432, Houston, TX 77030-4009, USA.
| | | | | |
Collapse
|
163
|
Higher levels of c-Met expression and phosphorylation identify cell lines with increased sensitivity to AMG-458, a novel selective c-Met inhibitor with radiosensitizing effects. Int J Radiat Oncol Biol Phys 2012; 84:e525-31. [PMID: 22836051 DOI: 10.1016/j.ijrobp.2012.06.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 06/11/2012] [Accepted: 06/15/2012] [Indexed: 01/19/2023]
Abstract
PURPOSE c-Met is overexpressed in some non-small cell lung cancer (NSCLC) cell lines and tissues. Cell lines with higher levels of c-Met expression and phosphorylation depend on this receptor for survival. We studied the effects of AMG-458 on 2 NSCLC cell lines. METHODS AND MATERIALS 3-(4,5-Dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assays assessed the sensitivities of the cells to AMG-458. Clonogenic survival assays illustrated the radiosensitizing effects of AMG-458. Western blot for cleaved caspase 3 measured apoptosis. Immunoblotting for c-Met, phospho-Met (p-Met), Akt/p-Akt, and Erk/p-Erk was performed to observe downstream signaling. RESULTS AMG-458 enhanced radiosensitivity in H441 but not in A549. H441 showed constitutive phosphorylation of c-Met. A549 expressed low levels of c-Met, which were phosphorylated only in the presence of exogenous hepatocyte growth factor. The combination of radiation therapy and AMG-458 treatment was found to synergistically increase apoptosis in the H441 cell line but not in A549. Radiation therapy, AMG-458, and combination treatment were found to reduce p-Akt and p-Erk levels in H441 but not in A549. H441 became less sensitive to AMG-458 after small interfering RNA knockdown of c-Met; there was no change in A549. After overexpression of c-Met, A549 became more sensitive, while H441 became less sensitive to AMG-458. CONCLUSIONS AMG-458 was more effective in cells that expressed higher levels of c-Met/p-Met, suggesting that higher levels of c-Met and p-Met in NSCLC tissue may classify a subset of tumors that are more sensitive to molecular therapies against this receptor.
Collapse
|
164
|
Hao GF, Yang GF, Zhan CG. Structure-based methods for predicting target mutation-induced drug resistance and rational drug design to overcome the problem. Drug Discov Today 2012; 17:1121-6. [PMID: 22789991 DOI: 10.1016/j.drudis.2012.06.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Revised: 06/01/2012] [Accepted: 06/29/2012] [Indexed: 11/15/2022]
Abstract
Drug resistance has become one of the biggest challenges in drug discovery and/or development and has attracted great research interests worldwide. During the past decade, computational strategies have been developed to predict target mutation-induced drug resistance. Meanwhile, various molecular design strategies, including targeting protein backbone, targeting highly conserved residues and dual/multiple targeting, have been used to design novel inhibitors for combating the drug resistance. In this article we review recent advances in development of computational methods for target mutation-induced drug resistance prediction and strategies for rational design of novel inhibitors that could be effective against the possible drug-resistant mutants of the target.
Collapse
Affiliation(s)
- Ge-Fei Hao
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, College of Chemistry, Central China Normal University, Wuhan 430079, PR China
| | | | | |
Collapse
|
165
|
Lee J, Han SY, Jung H, Yang J, Choi JW, Chae CH, Park CH, Choi SU, Lee K, Ha JD, Lee CO, Ryu JW, Kim HR, Koh JS, Cho SY. Synthesis and structure–activity relationship of aminopyridines with substituted benzoxazoles as c-Met kinase inhibitors. Bioorg Med Chem Lett 2012; 22:4044-8. [DOI: 10.1016/j.bmcl.2012.04.083] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 04/10/2012] [Accepted: 04/17/2012] [Indexed: 01/03/2023]
|
166
|
Logue JS, Morrison DK. Complexity in the signaling network: insights from the use of targeted inhibitors in cancer therapy. Genes Dev 2012; 26:641-50. [PMID: 22474259 DOI: 10.1101/gad.186965.112] [Citation(s) in RCA: 201] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer often arises when normal cellular growth goes awry due to defects in critical signal transduction pathways. A growing number of inhibitors that target specific components of these pathways are in clinical use, but the success of these agents has been limited by the resistance to inhibitor therapy that ultimately develops. Studies have now shown that cancer cells respond to chronic drug treatment by adapting their signaling circuitry, taking advantage of pathway redundancy and routes of feedback and cross-talk to maintain their function. This review focuses on the compensatory signaling mechanisms highlighted by the use of targeted inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Jeremy S Logue
- Laboratory of Cell and Developmental Signaling, National Cancer Institute at Frederick, Frederick, Maryland 21702, USA
| | | |
Collapse
|
167
|
Hepatocyte growth factor expression in EGFR mutant lung cancer with intrinsic and acquired resistance to tyrosine kinase inhibitors in a Japanese cohort. J Thorac Oncol 2012; 6:2011-7. [PMID: 22052230 DOI: 10.1097/jto.0b013e31823ab0dd] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION This study was performed to determine the incidence rates of resistance factors, i.e., high-level hepatocyte growth factor (HGF) expression, epidermal growth factor receptor (EGFR) T790M secondary mutation, and MET amplification, in tumors with intrinsic and acquired EGFR tyrosine kinase inhibitor (TKI) resistance in EGFR mutant lung cancer. METHODS Ninety-seven specimens from 93 EGFR mutant lung cancer patients (23 tumors with acquired resistance from 20 patients, 45 tumors with intrinsic resistance from 44 patients [nonresponders], 29 sensitive tumors from 29 patients) from 11 institutes in Japan were analyzed. HGF expression, EGFR T790M secondary mutation, and MET amplification were determined by immunohistochemistry, cycleave real-time polymerase chain reaction, and fluorescence in situ hybridization, respectively. RESULTS High-level HGF expression, EGFR T790M secondary mutation, and MET amplification were detected in 61, 52, and 9% of tumors with acquired resistance, respectively. High-level HGF expression was detected in 29% of tumors with intrinsic resistance (nonresponders), whereas EGFR T790M secondary mutation and MET amplification were detected in 0 and 4%, respectively. HGF expression was significantly higher in tumors with acquired resistance than in sensitive tumors (p < 0.001, Student's t test). Fifty percent of tumors with acquired resistance showed simultaneous HGF expression with EGFR T790M secondary mutation and MET amplification. CONCLUSIONS High-level HGF expression was detected more frequently than EGFR T790M secondary mutation or MET amplification in tumors with intrinsic and acquired EGFR-TKI resistance in EGFR mutant lung cancer in Japanese patients. These observations provide a rationale for targeting HGF in EGFR-TKI resistance in EGFR mutant lung cancer.
Collapse
|
168
|
Abstract
The majority of lung adenocarcinoma patients with epidermal growth factor receptor- (EGFR) mutated or EML4-ALK rearrangement-positive tumors are sensitive to tyrosine kinase inhibitors. Both primary and acquired resistance in a significant number of those patients to these therapies remains a major clinical problem. The specific molecular mechanisms associated with tyrosine kinase inhibitor resistance are not fully understood. Clinicopathological observations suggest that molecular alterations involving so-called 'driver mutations' could be used as markers that aid in the selection of patients most likely to benefit from targeted therapies. In this review, we summarize recent developments involving the specific molecular mechanisms and markers that have been associated with primary and acquired resistance to EGFR-targeted therapy in lung adenocarcinomas. Understanding these mechanisms may provide new treatment avenues and improve current treatment algorithms.
Collapse
|
169
|
Sennino B, Ishiguro-Oonuma T, Wei Y, Naylor RM, Williamson CW, Bhagwandin V, Tabruyn SP, You WK, Chapman HA, Christensen JG, Aftab DT, McDonald DM. Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov 2012; 2:270-87. [PMID: 22585997 DOI: 10.1158/2159-8290.cd-11-0240] [Citation(s) in RCA: 327] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
UNLABELLED Invasion and metastasis increase after the inhibition of VEGF signaling in some preclinical tumor models. In the present study we asked whether selective VEGF inhibition is sufficient to increase invasion and metastasis and whether selective c-Met inhibition is sufficient to block this effect. Treatment of pancreatic neuroendocrine tumors in RIP-Tag2 mice with a neutralizing anti-VEGF antibody reduced tumor burden but increased tumor hypoxia, hypoxia-inducible factor-1α, and c-Met activation and also increased invasion and metastasis. However, invasion and metastasis were reduced by concurrent inhibition of c-Met by PF-04217903 or PF-02341066 (crizotinib). A similar benefit was found in orthotopic Panc-1 pancreatic carcinomas treated with sunitinib plus PF-04217903 and in RIP-Tag2 tumors treated with XL184 (cabozantinib), which simultaneously blocks VEGF and c-Met signaling. These findings document that invasion and metastasis are promoted by selective inhibition of VEGF signaling and can be reduced by the concurrent inhibition of c-Met. SIGNIFICANCE This report examines the mechanism of increased tumor aggressiveness after anti-VEGF therapy and presents evidence for roles of vascular pruning, hypoxia, and c-Met activation. The results show that simultaneous inhibition of c-Met and VEGF signaling not only slows tumor growth but also reduces invasion and metastasis.
Collapse
Affiliation(s)
- Barbara Sennino
- Comprehensive Cancer Center, Department of Anatomy, University of California-San Francisco (UCSF), San Francisco, CA 94143-0452, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Tegze B, Szállási Z, Haltrich I, Pénzváltó Z, Tóth Z, Likó I, Gyorffy B. Parallel evolution under chemotherapy pressure in 29 breast cancer cell lines results in dissimilar mechanisms of resistance. PLoS One 2012; 7:e30804. [PMID: 22319589 PMCID: PMC3271089 DOI: 10.1371/journal.pone.0030804] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 12/21/2011] [Indexed: 11/18/2022] Open
Abstract
Background Developing chemotherapy resistant cell lines can help to identify markers of resistance. Instead of using a panel of highly heterogeneous cell lines, we assumed that truly robust and convergent pattern of resistance can be identified in multiple parallel engineered derivatives of only a few parental cell lines. Methods Parallel cell populations were initiated for two breast cancer cell lines (MDA-MB-231 and MCF-7) and these were treated independently for 18 months with doxorubicin or paclitaxel. IC50 values against 4 chemotherapy agents were determined to measure cross-resistance. Chromosomal instability and karyotypic changes were determined by cytogenetics. TaqMan RT-PCR measurements were performed for resistance-candidate genes. Pgp activity was measured by FACS. Results All together 16 doxorubicin- and 13 paclitaxel-treated cell lines were developed showing 2–46 fold and 3–28 fold increase in resistance, respectively. The RT-PCR and FACS analyses confirmed changes in tubulin isofom composition, TOP2A and MVP expression and activity of transport pumps (ABCB1, ABCG2). Cytogenetics showed less chromosomes but more structural aberrations in the resistant cells. Conclusion We surpassed previous studies by parallel developing a massive number of cell lines to investigate chemoresistance. While the heterogeneity caused evolution of multiple resistant clones with different resistance characteristics, the activation of only a few mechanisms were sufficient in one cell line to achieve resistance.
Collapse
Affiliation(s)
- Bálint Tegze
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary.
| | | | | | | | | | | | | |
Collapse
|
171
|
Abstract
Uncontrolled cell survival, growth, angiogenesis and metastasis are essential hallmarks of cancer. Genetic and biochemical data have demonstrated that the growth and motility factor hepatocyte growth factor/scatter factor (HGF/SF) and its receptor, the tyrosine kinase MET, have a causal role in all of these processes, thus providing a strong rationale for targeting these molecules in cancer. Parallel progress in understanding the structure and function of HGF/SF, MET and associated signalling components has led to the successful development of blocking antibodies and a large number of small-molecule MET kinase inhibitors. In this Review, we discuss these advances, as well as results from recent clinical studies that demonstrate that inhibiting MET signalling in several types of solid human tumours has major therapeutic value.
Collapse
Affiliation(s)
- Ermanno Gherardi
- Medical Research Council (MRC) Centre, Hills Road, Cambridge CB2 2QH, UK.
| | | | | | | |
Collapse
|
172
|
|
173
|
Lennerz JK, Kwak EL, Ackerman A, Michael M, Fox SB, Bergethon K, Lauwers GY, Christensen JG, Wilner KD, Haber DA, Salgia R, Bang YJ, Clark JW, Solomon BJ, Iafrate AJ. MET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinib. J Clin Oncol 2011; 29:4803-4810. [PMID: 22042947 PMCID: PMC3255989 DOI: 10.1200/jco.2011.35.4928] [Citation(s) in RCA: 359] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 06/20/2011] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Amplification of the MET proto-oncogene in gastroesophageal cancer (GEC) may constitute a molecular marker for targeted therapy. We examined a GEC cohort with follow-up and reported the clinical response of four additional patients with MET-amplified tumors to the small molecule inhibitor crizotinib as part of an expanded phase I cohort study. PATIENTS AND METHODS From 2007 to 2009, patients with GEC were genetically screened as a consecutive series of 489 tumors (stages 0, I, and II, 39%; III, 25%; IV, 36%; n = 222 esophageal, including n = 21 squamous carcinomas). MET, EGFR, and HER2 amplification status was assessed by using fluorescence in situ hybridization. RESULTS Ten (2%) of 489 patients screened harbored MET amplification; 23 (4.7%) harbored EGFR amplification; 45 (8.9%) harbored HER2 amplification; and 411 (84%) were wild type for all three genes (ie, negative). MET-amplified tumors were typically high-grade adenocarcinomas that presented at advanced stages (5%; n = 4 of 80). EGFR-amplified tumors showed the highest fraction of squamous cell carcinoma (17%; n = 4 of 23). HER2, MET, and EGFR amplification were, with one exception (MET and EGFR positive), mutually exclusive events. Survival analysis in patients with stages III and IV disease showed substantially shorter median survival in MET/EGFR-amplified groups, with a rank order for all groups by median survival (from most to least aggressive): MET (7.1 months; P < .001) less than EGFR (11.2 months; P = .16) less than HER2 (16.9 months; P = .89) when compared with the negative group (16.2 months). Two of four patients with MET-amplified tumors treated with crizotinib experienced tumor shrinkage (-30% and -16%) and experienced progression after 3.7 and 3.5 months. CONCLUSION MET amplification defines a small and aggressive subset of GEC with indications of transient sensitivity to the targeted MET inhibitor crizotinib (PF-02341066).
Collapse
Affiliation(s)
- Jochen K. Lennerz
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Eunice L. Kwak
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Allison Ackerman
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Michael Michael
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Stephen B. Fox
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Kristin Bergethon
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Gregory Y. Lauwers
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - James G. Christensen
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Keith D. Wilner
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Daniel A. Haber
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Ravi Salgia
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Yung-Jue Bang
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Jeffrey W. Clark
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - Benjamin J. Solomon
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| | - A. John Iafrate
- Jochen K. Lennerz, A. John Iafrate, Kristin Bergethon, Gregory Y. Lauwers, Daniel A. Haber, Eunice L. Kwak, Jeffrey W. Clark, Allison Ackerman, Massachusetts General Hospital/Harvard Medical School, Boston, MA; Michael Michael, Stephen B. Fox, Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Australia; James G. Christensen, Keith D. Wilner, Pfizer, La Jolla, CA; Ravi Salgia, University of Chicago, Chicago, IL; and Yung-Jue Bang, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
174
|
|
175
|
|
176
|
Abstract
Standard cytotoxic chemotherapy is effective for some cancers, but for many others, available treatments offer only a limited survival benefit. Lung adenocarcinoma is one such cancer, responsible for approximately half of lung cancer deaths each year. Development of targeted therapies is thought to hold the most promise for successfully treating this disease, but a targeted approach is dependent on understanding the genomic state of the tumor cells. Exon-directed sequencing of large numbers of lung adenocarcinoma tumor samples has provided an initial low-resolution image of the somatic mutation profile of these tumors. Such cancer sequencing studies have confirmed the high frequency of TP53 and KRAS mutations in lung adenocarcinoma, have found inactivating mutations in known tumor suppressor genes not previously associated with lung adenocarcinoma, and have identified oncogenic mutations of EGFR upon which the first targeted therapy for lung adenocarcinoma patients was based. Additional candidate oncogenes await functional validation. It is anticipated that upcoming whole-exome and whole-genome lung adenocarcinoma sequencing experiments will reveal a more detailed landscape of somatic mutations that can be exploited for therapeutic purposes.
Collapse
|
177
|
Potential therapeutic strategies to overcome acquired resistance to BRAF or MEK inhibitors in BRAF mutant cancers. Oncotarget 2011; 2:336-46. [PMID: 21505228 PMCID: PMC3248170 DOI: 10.18632/oncotarget.262] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent clinical trials with selective inhibitors of the BRAF and MEK kinases have shown promising results in patients with tumors harboring BRAF V600 mutations. However, as has been observed previously with similarly successful targeted therapies, acquired resistance to these agents is an emerging problem that limits their clinical benefit. Several recent studies from our laboratory and others have investigated the causes of acquired resistance to BRAF and MEK inhibitors, and multiple resistance mechanisms have been identified. Here, we review these mechanisms and suggest that they can be broadly grouped into two main classes: ERK-dependent and ERK-independent. We also propose distinct therapeutic strategies that might be employed to overcome each class of acquired resistance.
Collapse
|
178
|
Tiedt R, Degenkolbe E, Furet P, Appleton BA, Wagner S, Schoepfer J, Buck E, Ruddy DA, Monahan JE, Jones MD, Blank J, Haasen D, Drueckes P, Wartmann M, McCarthy C, Sellers WR, Hofmann F. A drug resistance screen using a selective MET inhibitor reveals a spectrum of mutations that partially overlap with activating mutations found in cancer patients. Cancer Res 2011; 71:5255-64. [PMID: 21697284 DOI: 10.1158/0008-5472.can-10-4433] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The emergence of drug resistance is a primary concern in any cancer treatment, including with targeted kinase inhibitors as exemplified by the appearance of Bcr-Abl point mutations in chronic myeloid leukemia (CML) patients treated with imatinib. In vitro approaches to identify resistance mutations in Bcr-Abl have yielded mutation spectra that faithfully recapitulated clinical observations. To predict resistance mutations in the receptor tyrosine kinase MET that could emerge during inhibitor treatment in patients, we conducted a resistance screen in BaF3 TPR-MET cells using the novel selective MET inhibitor NVP-BVU972. The observed spectrum of mutations in resistant cells was dominated by substitutions of tyrosine 1230 but also included other missense mutations and partially overlapped with activating MET mutations that were previously described in cancer patients. Cocrystallization of the MET kinase domain in complex with NVP-BVU972 revealed a key role for Y1230 in binding of NVP-BVU972, as previously reported for multiple other selective MET inhibitors. A second resistance screen in the same format with the MET inhibitor AMG 458 yielded a distinct spectrum of mutations rich in F1200 alterations, which is consistent with a different predicted binding mode. Our findings suggest that amino acid substitutions in the MET kinase domain of cancer patients need to be carefully monitored before and during treatment with MET inhibitors, as resistance may preexist or emerge. Compounds binding in the same manner as NVP-BVU972 might be particularly susceptible to the development of resistance through mutations in Y1230, a condition that may be addressed by MET inhibitors with alternative binding modes.
Collapse
MESH Headings
- Amino Acid Substitution
- Aminopyridines/metabolism
- Aminopyridines/pharmacology
- Animals
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Line, Transformed
- Cell Line, Tumor
- Crystallography, X-Ray
- DNA Mutational Analysis
- DNA, Neoplasm/genetics
- Drug Resistance, Neoplasm/genetics
- Enzyme Activation/genetics
- Humans
- Mice
- Models, Molecular
- Mutagenesis
- Mutation, Missense
- Neoplasms/drug therapy
- Neoplasms/genetics
- Point Mutation
- Protein Binding
- Protein Conformation
- Protein Kinase Inhibitors/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein Structure, Tertiary
- Proto-Oncogene Proteins c-met/antagonists & inhibitors
- Proto-Oncogene Proteins c-met/chemistry
- Proto-Oncogene Proteins c-met/genetics
- Pyrazoles/metabolism
- Pyrazoles/pharmacology
- Quinolines/metabolism
- Quinolines/pharmacology
- Receptors, Growth Factor/antagonists & inhibitors
- Receptors, Growth Factor/chemistry
- Receptors, Growth Factor/genetics
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Ralph Tiedt
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Atmaca A, Pauligk C, Steinmetz K, Altmannsberger HM, Jäger E, Al-Batran SE. Prognostic impact of phosphorylated mitogen-activated protein kinase expression in patients with metastatic gastric cancer. Oncology 2011; 80:130-4. [PMID: 21677458 DOI: 10.1159/000329063] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 04/15/2011] [Indexed: 12/24/2022]
Abstract
BACKGROUND This study was designed to investigate the expression of phosphorylated mitogen-activated protein kinase (p-MAPK/Erk1 and Erk2) and its correlation with outcomes in patients with metastatic gastric cancer. METHODS p-MAPK was detected by immunohistochemistry using monoclonal antibodies in a total of 223 formalin-fixed, paraffin-embedded samples obtained from 156 patients who received first-line chemotherapy in a phase III trial. RESULTS p-MAPK was positive in 93 (59.6%) and negative in 63 (40.4%) of the 156 patients evaluated. Similar rates of p-MAPK positivity were found in primaries (53%) and metastatic lesions (61.4%). Overall survival was significantly shorter in p-MAPK-positive patients (13.7 vs. 8.5 months) in the univariate analysis. However, this prognostic value disappeared as a trend in the multivariate analysis (p = 0.1). There was a strong, positive correlation between p-MAPK and the MIB-1 proliferation index, but MIB-1 did not predict outcomes. CONCLUSION p-MAPK expression could be a potential negative prognostic parameter in patients with metastatic gastric cancer treated with chemotherapy.
Collapse
Affiliation(s)
- Akin Atmaca
- Department of Hematology and Oncology, Krankenhaus Nordwest, Frankfurt am Main, Germany.
| | | | | | | | | | | |
Collapse
|
180
|
Katz JD, Jewell JP, Guerin DJ, Lim J, Dinsmore CJ, Deshmukh SV, Pan BS, Marshall CG, Lu W, Altman MD, Dahlberg WK, Davis L, Falcone D, Gabarda AE, Hang G, Hatch H, Holmes R, Kunii K, Lumb KJ, Lutterbach B, Mathvink R, Nazef N, Patel SB, Qu X, Reilly JF, Rickert KW, Rosenstein C, Soisson SM, Spencer KB, Szewczak AA, Walker D, Wang W, Young J, Zeng Q. Discovery of a 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one (MK-2461) inhibitor of c-Met kinase for the treatment of cancer. J Med Chem 2011; 54:4092-108. [PMID: 21608528 DOI: 10.1021/jm200112k] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
c-Met is a transmembrane tyrosine kinase that mediates activation of several signaling pathways implicated in aggressive cancer phenotypes. In recent years, research into this area has highlighted c-Met as an attractive cancer drug target, triggering a number of approaches to disrupt aberrant c-Met signaling. Screening efforts identified a unique class of 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one kinase inhibitors, exemplified by 1. Subsequent SAR studies led to the development of 81 (MK-2461), a potent inhibitor of c-Met that was efficacious in preclinical animal models of tumor suppression. In addition, biochemical studies and X-ray analysis have revealed that this unique class of kinase inhibitors binds preferentially to the activated (phosphorylated) form of the kinase. This report details the development of 81 and provides a description of its unique biochemical properties.
Collapse
Affiliation(s)
- Jason D Katz
- Department of Chemistry, Merck Research Laboratories, 33 Avenue Louis Pasteur, BMB-2-114, Boston, Massachusetts 02115, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|