101
|
Godar M, Morello V, Sadi A, Hultberg A, De Jonge N, Basilico C, Hanssens V, Saunders M, Lambrecht BN, El Khattabi M, de Haard H, Michieli P, Blanchetot C. Dual anti-idiotypic purification of a novel, native-format biparatopic anti-MET antibody with improved in vitro and in vivo efficacy. Sci Rep 2016; 6:31621. [PMID: 27546726 PMCID: PMC4992859 DOI: 10.1038/srep31621] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/22/2016] [Indexed: 12/15/2022] Open
Abstract
Bispecific antibodies are of great interest due to their ability to simultaneously bind and engage different antigens or epitopes. Nevertheless, it remains a challenge to assemble, produce and/or purify them. Here we present an innovative dual anti-idiotypic purification process, which provides pure bispecific antibodies with native immunoglobulin format. Using this approach, a biparatopic IgG1 antibody targeting two distinct, HGF-competing, non-overlapping epitopes on the extracellular region of the MET receptor, was purified with camelid single-domain antibody fragments that bind specifically to the correct heavy chain/light chain pairings of each arm. The purity and functionality of the anti-MET biparatopic antibody was then confirmed by mass spectrometry and binding experiments, demonstrating its ability to simultaneously target the two epitopes recognized by the parental monoclonal antibodies. The improved MET-inhibitory activity of the biparatopic antibody compared to the parental monoclonal antibodies, was finally corroborated in cell-based assays and more importantly in a tumor xenograft mouse model. In conclusion, this approach is fast and specific, broadly applicable and results in the isolation of a pure, novel and native-format anti-MET biparatopic antibody that shows superior biological activity over the parental monospecific antibodies both in vitro and in vivo.
Collapse
Affiliation(s)
- Marie Godar
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium.,VIB Inflammation Research Center 9052 Zwijnaarde, Belgium.,Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium
| | - Virginia Morello
- Department of Oncology, University of Torino Medical School, 10060 Candiolo, Turin, Italy.,Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Turin, Italy
| | - Ava Sadi
- QVQ BV, Yalelaan 1 Androclus building, 3584 CL Utrecht, The Netherlands
| | - Anna Hultberg
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium
| | - Natalie De Jonge
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium
| | | | - Valérie Hanssens
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium
| | - Michael Saunders
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium
| | - Bart N Lambrecht
- VIB Inflammation Research Center 9052 Zwijnaarde, Belgium.,Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium.,Department of Pulmonary Medicine, ErasmusMC, 3015 GE Rotterdam, The Netherlands
| | | | - Hans de Haard
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium
| | - Paolo Michieli
- Department of Oncology, University of Torino Medical School, 10060 Candiolo, Turin, Italy.,Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Turin, Italy
| | | |
Collapse
|
102
|
Dual MET/EGFR therapy leads to complete response and resistance prevention in a MET-amplified gastroesophageal xenopatient cohort. Oncogene 2016; 36:1200-1210. [PMID: 27524418 DOI: 10.1038/onc.2016.283] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/10/2016] [Accepted: 06/23/2016] [Indexed: 12/12/2022]
Abstract
Amplification of the MET oncogene occurs in 2-4% of gastroesophageal cancers and defines a small and aggressive subset of tumors. Although in vitro studies have given very promising results, clinical trials with MET inhibitors have been disappointing, showing few and short lasting responses. The aim of the work was to exploit a MET-amplified patient-derived xenograft model to optimize anti-MET therapeutic strategies in gastroesophageal cancer. We found that despite the high MET amplification level (26 gene copies), in the absence of qualitative or quantitative alterations of EGFR, MET inhibitors induced only tumor growth inhibition, whereas dual MET/EGFR inhibition led to complete tumor regression. Importantly, the combo treatment completely prevented the onset of resistance, which quite rapidly appeared in tumors treated with MET monotherapy. We found that this secondary resistance was due to EGFR activation and could be overcome by dual MET/EGFR inhibition. Similar results were also obtained in a MET-addicted, established gastric cancer cell line. In vitro experiments performed on tumor-derived primary cells confirmed that MET inhibitors were not able to abrogate the activation of downstream transducers and that only the combined MET/EGFR treatment completely shut off the signaling. Previously reported cases, as well as those described here, showed only partial and transient sensitivity to anti-MET therapy. The finding that combined anti-MET/EGFR therapy-even in the absence of EGFR genetic alterations-induced complete and durable response, represents a proof of concept and guarantees further investigations, opening a new perspective of treatment for these patients.
Collapse
|
103
|
Zhang Z, Kong Y, Yang W, Ma F, Zhang Y, Ji S, Ma EM, Liu H, Chen Y, Hua Y. Upregulation of microRNA-34a enhances the DDP sensitivity of gastric cancer cells by modulating proliferation and apoptosis via targeting MET. Oncol Rep 2016; 36:2391-7. [PMID: 27513895 DOI: 10.3892/or.2016.5016] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/14/2016] [Indexed: 12/15/2022] Open
Abstract
Cisplatin (DDP) based chemotherapy is still the main strategy of human gastric cancer (GC) treatment. However, drug resistance is a major obstacle for DDP chemotherapy. Recent studies indicated that the resistance could be modulated by the regulation of dysregulated microRNAs (miRs). Previous study also found miR-34a was associated with cell proliferation and apoptosis in human GC; however, the relationship between miR-34a and DDP resistance still remains unexplored. The purpose of this study was to investigate whether miR-34a is associated with DDP resistance in human GC cells. Our study found that the expression of miR-34a was significantly decreased in DDP resistance human GC tissues and DDP resistance human GC SGC7901/DDP cells compared with normal GC tissues and cells. Upregulation of miR-34a enhanced the DDP sensitivity of SGC7901/DDP cells to DDP through the inhibition of cell proliferation and induction of cell apoptosis; on the other hand downregulation of miR-34a could weaken the DDP sensitivity of SGC7901 cells to DDP. Further study found that MET was a direct target of miR-34a and the regulation of MET could affect the DDP sensitivity of SGC7901/DDP cells. Moreover, our study also indicated that up-regulation of miR-34a could decrease the expression of MET in SGC7901/DDP cells. Therefore, our findings suggested miR-34a could modulate human gastric cancer cell DDP sensitivity by regulation of cell proliferation and apoptosis via targeting MET, potentially benefiting human GC treatment in the future.
Collapse
Affiliation(s)
- Zhandong Zhang
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Ye Kong
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Wei Yang
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Fei Ma
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Yonglei Zhang
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Sheqing Ji
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Er-Min Ma
- Surgical Oncology, The People Hospital of Zhengzhou, Zhengzhou, Henan 450000, P.R. China
| | - Hongxing Liu
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Yongshun Chen
- Department of Radiotherapy, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Yawei Hua
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| |
Collapse
|
104
|
Zheng D, Wang R, Ye T, Yu S, Hu H, Shen X, Li Y, Ji H, Sun Y, Chen H. MET exon 14 skipping defines a unique molecular class of non-small cell lung cancer. Oncotarget 2016; 7:41691-41702. [PMID: 27223439 PMCID: PMC5173088 DOI: 10.18632/oncotarget.9541] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/08/2016] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Recurrent MET exon 14 splicing has been revealed in lung cancers and is a promising therapeutic target. Because we have limited knowledge about the natural history of MET mutant tumors, the current study was aiming to determine the clinical and pathological characteristics in non-small cell lung cancers (NSCLC). RESULTS Twenty-three patients (1.3%) were positive for MET exon 14 skipping. Patients with MET exon 14 skipping displayed unique characteristics: female, non-smokers, earlier pathology stage and older age. MET exon 14 skipping indicated an early event as other drivers in lung cancer, while MET copy number gain was more likely a late event in lung cancer. Overall survival (OS) of patients harboring MET exon 14 skipping was longer than patients with KRAS mutation. Almost four-fifths of the lung tumors with MET exon 14 skipping had EGFR and/or HER2 gene copy number gains. EGFR inhibitor showed moderate antitumor activity in treatment of a patient harboring MET exon 14 skipping. PATIENTS AND METHODS From October 2007 to June 2013, we screened 1770 patients with NSCLC and correlated MET status with clinical pathologic characteristics and mutations in EGFR, KRAS, BRAF, HER2, and ALK. Quantitative Real-Time PCR was used to detect MET gene copy number gain. Immunohistochemistry (IHC) was also performed to screen MET exon 14 skipping. Clinicopathological characteristics and survival information were analyzed. CONCLUSIONS MET exon 14 skipping was detected in 1.3% (23/1770) of the Chinese patients with NSCLC. MET exon 14 skipping defined a new molecular subset of NSCLC with identifiable clinical characteristics. The therapeutic EGFR inhibitors might be an alternative treatment for patients with MET mutant NSCLC.
Collapse
Affiliation(s)
- Difan Zheng
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rui Wang
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ting Ye
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Su Yu
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Cancer Research Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Haichuan Hu
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Xuxia Shen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yuan Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hongbin Ji
- Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, China
| | - Yihua Sun
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haiquan Chen
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
105
|
Pupo E, Ducano N, Lupo B, Vigna E, Avanzato D, Perera T, Trusolino L, Lanzetti L, Comoglio PM. Rebound Effects Caused by Withdrawal of MET Kinase Inhibitor Are Quenched by a MET Therapeutic Antibody. Cancer Res 2016; 76:5019-29. [DOI: 10.1158/0008-5472.can-15-3107] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 06/05/2016] [Indexed: 11/16/2022]
|
106
|
Zhang Y, Du Z, Zhang M. Biomarker development in MET-targeted therapy. Oncotarget 2016; 7:37370-37389. [PMID: 27013592 PMCID: PMC5095083 DOI: 10.18632/oncotarget.8276] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 03/14/2016] [Indexed: 12/16/2022] Open
Abstract
Activation of the MET receptor tyrosine kinase by its ligand, hepatocyte growth factor (HGF), has been implicated in a variety of cellular processes, including cell proliferation, survival, migration, motility and invasion, all of which may be enhanced in human cancers. Aberrantly activated MET/HGF signaling correlates with tumorigenesis and metastasis, and is regarded as a robust target for the development of novel anti-cancer treatments. Various clinical trials were conducted to evaluate the safety and efficacy of selective HGF/MET inhibitors in cancer patients. There is currently no optimal or standardized method for accurate and reliable assessment of MET levels, or other biomarkers that are predictive of the patient response to MET-targeted therapeutics. In this review, we discuss the importance of accurate HGF/MET signal detection as a predictive biomarker to guide patient selection for clinical trials of MET-targeted therapies in human cancers.
Collapse
Affiliation(s)
- Yanni Zhang
- Amgen Biopharmaceutical Research and Development (Shanghai) Co., Ltd, Shanghai, China
| | - Zhiqiang Du
- Amgen Biopharmaceutical Research and Development (Shanghai) Co., Ltd, Shanghai, China
| | - Mingqiang Zhang
- Amgen Biopharmaceutical Research and Development (Shanghai) Co., Ltd, Shanghai, China
| |
Collapse
|
107
|
c-Met expression and MET amplification in malignant pleural mesothelioma. Ann Diagn Pathol 2016; 23:1-7. [PMID: 27402216 DOI: 10.1016/j.anndiagpath.2016.04.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 04/13/2016] [Indexed: 01/04/2023]
Abstract
c-Met is a receptor tyrosine kinase shown to be overexpressed in malignant pleural mesothelioma (MPM). Whereas MET mutations have been identified in 3%-16% of MPMs, MET amplification has recently been reported in a single epithelioid MPM. We studied c-Met expression and MET amplification in a large MPM cohort and correlated results with morphologic and clinical features. We report the first case of MET amplification in sarcomatoid MPM. MPMs from surgical pathology files (1989-2014) were reviewed. c-Met immunohistochemistry was performed. Staining intensity and distribution were multiplied (H-score). Staining localization (cytoplasmic and/or membranous) was noted. Fluorescence in situ hybridization was performed using probes for MET and centromere 7. One hundred forty-nine patients (median age, 68.0years; interquartile range, 61-75) had epithelioid (n=97), biphasic (n=18), or sarcomatoid (n=34) MPM. Median follow-up was 10.1months (range, 0.1-222.5). One hundred thirty patients died of disease; 2 were alive with disease. c-Met was expressed in 147 MPMs. c-Met staining intensity, distribution, and H-score differed among the histologic subtypes (P=.015; P=.0001, and P=.0005, respectively), but none were predictive of survival. Epithelioid subtype had greater c-Met expression. MET amplification was identified in 1 sarcomatoid MPM and MET duplication in 1 epithelioid MPM; both had poor outcomes. Chromosome 7 aneusomy was observed in 54 of 144 (37.5%) MPMs and associated with decreased overall survival in sarcomatoid MPMs (hazard ratio=2.81; 95% confidence interval, 1.21-6.51; P=.01). In conclusion, c-Met is expressed in MPM, with significant differences in expression among histologic subtypes. MET amplification is a rare event in MPM, making it an unlikely common pathogenesis for c-Met expression.
Collapse
|
108
|
An N, Xiong Y, LaRue AC, Kraft AS, Cen B. Activation of Pim Kinases Is Sufficient to Promote Resistance to MET Small-Molecule Inhibitors. Cancer Res 2016; 75:5318-28. [PMID: 26670562 DOI: 10.1158/0008-5472.can-15-0544] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesenchymal-epithelial transition (MET) blockade offers a new targeted therapy particularly in those cancers with MET amplification. However, the efficacy and the duration of the response to MET inhibitors are limited by the emergence of drug resistance. Here, we report that resistance to small-molecule inhibitors of MET can arise from increased expression of the prosurvival Pim protein kinases. This resistance mechanism was documented in non-small cell lung cancer and gastric cancer cells with MET amplification. Inhibition of Pim kinases enhanced cell death triggered by short-term treatment with MET inhibitors. Pim kinases control the translation of antiapoptotic protein Bcl-2 at an internal ribosome entry site and this mechanism was identified as the basis for Pim-mediated resistance to MET inhibitors. Protein synthesis was increased in drug-resistant cells, secondary to a Pim-mediated increase in cap-independent translation. In cells rendered drug resistant by chronic treatment with MET inhibitors, genetic or pharmacologic inhibition of Pim kinases was sufficient to restore sensitivity in vitro and in vivo. Taken together, our results rationalize Pim inhibition as a strategy to augment responses and blunt acquired resistance to MET inhibitors in cancer.
Collapse
Affiliation(s)
- Ningfei An
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina. The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Ying Xiong
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Amanda C LaRue
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina. Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | | | - Bo Cen
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina. The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
109
|
Non-canonical dynamic mechanisms of interaction between the p66Shc protein and Met receptor. Biochem J 2016; 473:1617-27. [PMID: 27048591 PMCID: PMC4888465 DOI: 10.1042/bcj20160249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/04/2016] [Indexed: 11/26/2022]
Abstract
The present study identifies a novel and unexpected mechanism underscoring the diversification of p66Shc among other Shc (Src homology and collagen homology) proteins, with respect to its mode of interaction with the receptor Met and impacts on key binding effectors of Met-regulated signalling. Met receptor tyrosine kinase (RTK) is known to bind to the three distinct protein isoforms encoded by the ShcA (Shc) gene. Structure–function studies have unveiled critical roles for p52Shc-dependent signalling pathways in Met-regulated biological functions. The molecular basis of the interaction between the Met and p52Shc proteins is well-defined, but not for the longest protein isoform, p66Shc. In the present study, co-immunoprecipitation assays were performed in human embryonic kidney 293 (HEK293) cells, transiently co-transfected with Met and p66Shc mutants, in order to define the molecular determinants involved in mediating Met–p66Shc interaction. Our results show that p66Shc interacts constitutively with the receptor Met, and the Grb2 (growth factor receptor-bound protein-2) and Gab1 (Grb2-associated binder-1) adaptor proteins. Although its phosphotyrosine-binding domain (PTB) and Src homology 2 (SH2) domains co-ordinate p66Shc binding to non-activated Met receptor, these phosphotyrosine-binding modules, and its collagen homology domain 2 (CH2) region, exert negative constraints. In contrast, p66Shc interaction with the activated Met depends mainly on the integrity of its PTB domain, and to a lesser extent of its SH2 domain. Even though not required for the recruitment of p66Shc, tyrosine phosphorylation of p66Shc by activated Met enhances these interactions by mechanisms not reliant on the integrity of the Met multisubstrate-binding site. In turn, this increases phosphotyrosine-dependent p66Shc–Grb2–Gab1 complex formation away from the receptor, while blocking Grb2 and Gab1 recruitment to activated Met. In conclusion, we identify, for the first time, a novel non-canonical dynamic mode of interaction between Met and the p66 protein isoform of Shc and its effects on rewiring binding effector complexes according to the activation state of the receptor.
Collapse
|
110
|
Cignetto S, Modica C, Chiriaco C, Fontani L, Milla P, Michieli P, Comoglio PM, Vigna E. Dual Constant Domain-Fab: A novel strategy to improve half-life and potency of a Met therapeutic antibody. Mol Oncol 2016; 10:938-48. [PMID: 27103110 DOI: 10.1016/j.molonc.2016.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/01/2016] [Accepted: 03/20/2016] [Indexed: 12/12/2022] Open
Abstract
The kinase receptor encoded by the Met oncogene is a sensible target for cancer therapy. The chimeric monovalent Fab fragment of the DN30 monoclonal antibody (MvDN30) has an odd mechanism of action, based on cell surface removal of Met via activation of specific plasma membrane proteases. However, the short half-life of the Fab, due to its low molecular weight, is a severe limitation for the deployment in therapy. This issue was addressed by increasing the Fab molecular weight above the glomerular filtration threshold through the duplication of the constant domains, in tandem (DCD-1) or reciprocally swapped (DCD-2). The two newly engineered molecules showed biochemical properties comparable to the original MvDN30 in vitro, acting as full Met antagonists, impairing Met phosphorylation and activation of downstream signaling pathways. As a consequence, Met-mediated biological responses were inhibited, including anchorage-dependent and -independent cell growth. In vivo DCD-1 and DCD-2 showed a pharmacokinetic profile significantly improved over the original MvDN30, doubling the circulating half-life and reducing the clearance. In pre-clinical models of cancer, generated by injection of tumor cells or implant of patient-derived samples, systemic administration of the engineered molecules inhibited the growth of Met-addicted tumors.
Collapse
Affiliation(s)
- Simona Cignetto
- Candiolo Cancer Institute, FPO-IRCCS, Str Prov 142, 10060 Candiolo, Italy; University of Turin, Department of Oncology, Str Prov 142, 10060 Candiolo, Italy
| | - Chiara Modica
- Candiolo Cancer Institute, FPO-IRCCS, Str Prov 142, 10060 Candiolo, Italy; University of Turin, Department of Oncology, Str Prov 142, 10060 Candiolo, Italy
| | - Cristina Chiriaco
- Candiolo Cancer Institute, FPO-IRCCS, Str Prov 142, 10060 Candiolo, Italy
| | - Lara Fontani
- Candiolo Cancer Institute, FPO-IRCCS, Str Prov 142, 10060 Candiolo, Italy
| | - Paola Milla
- University of Turin, Department of Science and Drug Technology, Via P. Giuria 9, 10125 Turin, Italy
| | - Paolo Michieli
- Candiolo Cancer Institute, FPO-IRCCS, Str Prov 142, 10060 Candiolo, Italy; University of Turin, Department of Oncology, Str Prov 142, 10060 Candiolo, Italy
| | - Paolo M Comoglio
- Candiolo Cancer Institute, FPO-IRCCS, Str Prov 142, 10060 Candiolo, Italy; University of Turin, Department of Oncology, Str Prov 142, 10060 Candiolo, Italy.
| | - Elisa Vigna
- Candiolo Cancer Institute, FPO-IRCCS, Str Prov 142, 10060 Candiolo, Italy; University of Turin, Department of Oncology, Str Prov 142, 10060 Candiolo, Italy.
| |
Collapse
|
111
|
Nishimura Y, Hyuga S, Takiguchi S, Hyuga M, Itoh K, Hanawa T. Ephedrae herba stimulates hepatocyte growth factor-induced MET endocytosis and downregulation via early/late endocytic pathways in gefitinib-resistant human lung cancer cells. Int J Oncol 2016; 48:1895-906. [PMID: 26983447 DOI: 10.3892/ijo.2016.3426] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/23/2015] [Indexed: 11/06/2022] Open
Abstract
The MET tyrosine kinase receptor and its ligand, hepatocyte growth factor (HGF), are known to be overexpressed in a variety of malignant tumor cells, and are implicated in the development of gefitinib-resistance in human non-small cell lung cancer (NSCLC) cells. Ephedrae herba was previously reported to prevent HGF-induced cancer cell motility by directly suppressing HGF/MET signaling through the inhibition of MET tyrosine kinase, and treatment with its extract also considerably reduced MET protein levels. To further investigate the mechanism underlying the Ephedrae herba-induced inhibition of MET phosphorylation as well as its degradation and subsequent disappearance, we examined the effect of Ephedrae herba on HGF-stimulated MET endocytosis and downregulation via early/late endocytic pathways in an NSCLC cell line. Using immunofluorescence microscopy, we found that pretreatment of cells with Ephedrae herba extract dramatically changed the intracellular distribution of plasma membrane-associated MET, and that the resultant MET staining was distributed throughout the cytoplasm. Pretreatment of the cells with Ephedrae herba extract also led to the rapid loss of MET and phosphorylated (p)-MET in HGF-stimulated cells. In contrast, inefficient endocytic delivery of MET and p-MET from early to late endosomes was observed in the absence of Ephedrae herba extract, since considerable amounts of the internalized MET accumulated in the early endosomes and were not delivered to lysosomes up to 1 h after HGF-stimulation. Furthermore, large amounts of MET and p-MET that had accumulated in late endosomes of Ephedrae herba-pretreated cells after HGF stimulation were observed along with bafilomycin A1. Therefore, we inferred that degradation of MET occurred in the late endosome/lysosome pathway. Moreover, western blot analysis revealed the accelerated degradation of MET and p-MET proceeds in cells pretreated with Ephedrae herba extract. Collectively, our results suggest that some components of Ephedrae herba have a novel role in promoting HGF-stimulated MET and p-MET endocytosis followed by its downregulation, likely mediated by the early/late endocytic pathways.
Collapse
Affiliation(s)
- Yukio Nishimura
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Sumiko Hyuga
- Department of Clinical Research, Oriental Medicine Research Center of Kitasato University, Tokyo 108-8642, Japan
| | - Soichi Takiguchi
- Institute for Clinical Research, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Masashi Hyuga
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Kazuyuki Itoh
- Department of Biology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka 537-8511, Japan
| | - Toshihiko Hanawa
- Department of Clinical Research, Oriental Medicine Research Center of Kitasato University, Tokyo 108-8642, Japan
| |
Collapse
|
112
|
Tong JH, Yeung SF, Chan AWH, Chung LY, Chau SL, Lung RWM, Tong CY, Chow C, Tin EKY, Yu YH, Li H, Pan Y, Chak WP, Ng CSH, Mok TSK, To KF. MET Amplification and Exon 14 Splice Site Mutation Define Unique Molecular Subgroups of Non-Small Cell Lung Carcinoma with Poor Prognosis. Clin Cancer Res 2016; 22:3048-56. [PMID: 26847053 DOI: 10.1158/1078-0432.ccr-15-2061] [Citation(s) in RCA: 347] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/16/2016] [Indexed: 12/12/2022]
Abstract
PURPOSE Activation of MET oncogene as the result of amplification or activation mutation represents an emerging molecular target for cancer treatment. We comprehensively studied MET alterations and the clinicopathologic correlations in a large cohort of treatment-naïve non-small cell lung carcinoma (NSCLC). EXPERIMENTAL DESIGN Six hundred eighty-seven NSCLCs were tested for MET exon 14 splicing site mutation (METΔ14), DNA copy number alterations, and protein expression by Sanger sequencing, FISH, and IHC, respectively. RESULTS METΔ14 mutation was detected in 2.62% (18/687) of NSCLC. The mutation rates were 2.6% in adenocarcinoma, 4.8% in adenosquamous carcinoma, and 31.8% in sarcomatoid carcinoma. METΔ14 mutation was not detected in squamous cell carcinoma, large cell carcinoma, and lymphoepithelioma-like carcinoma but significantly enriched in sarcomatoid carcinoma (P < 0.001). METΔ14 occurred mutually exclusively with known driver mutations but tended to coexist with MET amplification or copy number gain (P < 0.001). Low-level MET amplification and polysomy might occur in the background of EGFR or KRAS mutation whereas high-level amplification (MET/CEP7 ratio ≥5) was mutually exclusive to the major driver genes except METΔ14. Oncogenic METΔ14 mutation and/or high-level amplification occurred in a total of 3.3% (23/687) of NSCLC and associated with higher MET protein expression. METΔ14 occurred more frequently in older patients whereas amplification was more common in ever-smokers. Both METΔ14 and high-level amplification were independent prognostic factors that predicted poorer survival by multivariable analysis. CONCLUSIONS The high incidence of METΔ14 mutation in sarcomatoid carcinoma suggested that MET inhibition might benefit this specific subgroup of patients. Clin Cancer Res; 22(12); 3048-56. ©2016 AACRSee related commentary by Drilon, p. 2832.
Collapse
Affiliation(s)
- Joanna H Tong
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Sai F Yeung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Anthony W H Chan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Lau Y Chung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Shuk L Chau
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Raymond Wai Ming Lung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Carol Y Tong
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Chit Chow
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Edith K Y Tin
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Yau H Yu
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Hui Li
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Yi Pan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Wing P Chak
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Calvin S H Ng
- Division of Cardiothoracic Surgery, Department of Surgery, The Chinese University of Hong Kong, Hong Kong
| | - Tony S K Mok
- Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong. Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong
| | - Ka F To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong. State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong. Li Ka-Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
113
|
Park CH, Cho SY, Ha JD, Jung H, Kim HR, Lee CO, Jang IY, Chae CH, Lee HK, Choi SU. Novel c-Met inhibitor suppresses the growth of c-Met-addicted gastric cancer cells. BMC Cancer 2016; 16:35. [PMID: 26801760 PMCID: PMC4722623 DOI: 10.1186/s12885-016-2058-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 01/10/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND c-Met signaling has been implicated in oncogenesis especially in cells with c-met gene amplification. Since 20 % of gastric cancer patients show high level of c-Met expression, c-Met has been identified as a good candidate for targeted therapy in gastric cancer. Herein, we report our newly synthesized c-Met inhibitor by showing its efficacy both in vitro and in vivo. METHODS Compounds with both triazolopyrazine and pyridoxazine scaffolds were synthesized and tested using HTRF c-Met kinase assay. We performed cytotoxic assay, cellular phosphorylation assay, and cell cycle assay to investigate the cellular inhibitory mechanism of our compounds. We also conducted mouse xenograft assay to see efficacy in vivo. RESULTS KRC-00509 and KRC-00715 were selected as excellent c-Met inhibitors through biochemical assay, and exhibited to be exclusively selective to c-Met by kinase panel assay. Cytotoxic assays using 18 gastric cancer cell lines showed our c-Met inhibitors suppressed specifically the growth of c-Met overexpressed cell lines, not that of c-Met low expressed cell lines, by inducing G1/S arrest. In c-met amplified cell lines, c-Met inhibitors reduced the downstream signals including Akt and Erk as well as c-Met activity. In vivo Hs746T xenograft assay showed KRC-00715 reduced the tumor size significantly. CONCLUSIONS Our in vitro and in vivo data suggest KRC-00715 is a potent and highly selective c-Met inhibitor which may have therapeutic potential in gastric tumor with c-Met overexpression.
Collapse
Affiliation(s)
- Chi Hoon Park
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon, 305-600, Republic of Korea.,Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon, 305-350, Republic of Korea
| | - Sung Yun Cho
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon, 305-600, Republic of Korea.,Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon, 305-350, Republic of Korea
| | - Jae Du Ha
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon, 305-600, Republic of Korea
| | - Heejung Jung
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon, 305-600, Republic of Korea.,Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon, 305-350, Republic of Korea
| | - Hyung Rae Kim
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon, 305-600, Republic of Korea
| | - Chong Ock Lee
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon, 305-600, Republic of Korea
| | - In-Young Jang
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon, 305-600, Republic of Korea
| | - Chong Hak Chae
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon, 305-600, Republic of Korea
| | - Heung Kyoung Lee
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon, 305-600, Republic of Korea
| | - Sang Un Choi
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Daejeon, 305-600, Republic of Korea.
| |
Collapse
|
114
|
Dai L, Trillo-Tinoco J, Cao Y, Bonstaff K, Doyle L, Del Valle L, Whitby D, Parsons C, Reiss K, Zabaleta J, Qin Z. Targeting HGF/c-MET induces cell cycle arrest, DNA damage, and apoptosis for primary effusion lymphoma. Blood 2015; 126:2821-31. [PMID: 26531163 PMCID: PMC4692142 DOI: 10.1182/blood-2015-07-658823] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/03/2015] [Indexed: 11/20/2022] Open
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV) is a principal causative agent of primary effusion lymphoma (PEL) with a poor prognosis in immunocompromised patients. However, it still lacks effective treatment which urgently requires the identification of novel therapeutic targets for PEL. Here, we report that the hepatocyte growth factor (HGF)/c-MET pathway is highly activated by KSHV in vitro and in vivo. The selective c-MET inhibitor, PF-2341066, can induce PEL apoptosis through cell cycle arrest and DNA damage, and suppress tumor progression in a xenograft murine model. By using microarray analysis, we identify many novel genes that are potentially controlled by HGF/c-MET within PEL cells. One of the downstream candidates, ribonucleoside-diphosphate reductase subunit M2 (RRM2), also displays the promising therapeutic value for PEL treatment. Our findings provide the framework for development of HGF/c-MET-focused therapy and implementation of clinical trials for PEL patients.
Collapse
Affiliation(s)
- Lu Dai
- Department of Microbiology, Immunology, & Parasitology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA; Research Center for Translational Medicine and Key Laboratory of Arrhythmias, East Hospital, Tongji University School of Medicine, Shanghai, China; Department of Medicine and
| | - Jimena Trillo-Tinoco
- Department of Pathology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA
| | - Yueyu Cao
- Research Center for Translational Medicine and Key Laboratory of Arrhythmias, East Hospital, Tongji University School of Medicine, Shanghai, China
| | | | | | - Luis Del Valle
- Department of Pathology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA
| | - Denise Whitby
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD; and
| | | | | | - Jovanny Zabaleta
- Department of Pediatrics, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA
| | - Zhiqiang Qin
- Department of Microbiology, Immunology, & Parasitology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA; Research Center for Translational Medicine and Key Laboratory of Arrhythmias, East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
115
|
Aptamers Binding to c-Met Inhibiting Tumor Cell Migration. PLoS One 2015; 10:e0142412. [PMID: 26658271 PMCID: PMC4676636 DOI: 10.1371/journal.pone.0142412] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 10/21/2015] [Indexed: 01/04/2023] Open
Abstract
The human receptor tyrosine kinase c-Met plays an important role in the control of critical cellular processes. Since c-Met is frequently over expressed or deregulated in human malignancies, blocking its activation is of special interest for therapy. In normal conditions, the c-Met receptor is activated by its bivalent ligand hepatocyte growth factor (HGF). Also bivalent antibodies can activate the receptor by cross linking, limiting therapeutic applications. We report the generation of the RNA aptamer CLN64 containing 2'-fluoro pyrimidine modifications by systematic evolution of ligands by exponential enrichment (SELEX). CLN64 and a previously described single-stranded DNA (ssDNA) aptamer CLN3 exhibited high specificities and affinities to recombinant and cellular expressed c-Met. Both aptamers effectively inhibited HGF-dependent c-Met activation, signaling and cell migration. We showed that these aptamers did not induce c-Met activation, revealing an advantage over bivalent therapeutic molecules. Both aptamers were shown to bind overlapping epitopes but only CLN3 competed with HGF binding to cMet. In addition to their therapeutic and diagnostic potential, CLN3 and CLN64 aptamers exhibit valuable tools to further understand the structural and functional basis for c-Met activation or inhibition by synthetic ligands and their interplay with HGF binding.
Collapse
|
116
|
Gray AL, Coleman DT, Castore RF, Mohyeldin MM, El Sayed KA, Cardelli JA. Isothiocyanatostilbenes as novel c-Met inhibitors. Oncotarget 2015; 6:41180-93. [PMID: 26543230 PMCID: PMC4747398 DOI: 10.18632/oncotarget.5748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 09/17/2015] [Indexed: 01/17/2023] Open
Abstract
The hepatocyte growth factor receptor (HGFR or c-Met) is a driver of multiple cancer subtypes. While there are several c-Met inhibitors in development, few have been approved for clinical use, warranting the need for continued research and development of c-Met targeting therapeutic modalities. The research presented here demonstrates a particular class of compounds known as isothiocyanatostilbenes can act as c-Met inhibitors in multiple cancer cell lines. Specifically, we found that 4,4′-Diisothiocyanatostilbene-2,2′-disulfonic acid (DIDS) and 4,4′-Diisothiocyanatodihydrostilbene-2,2′-disulfonic acid (H2DIDS) had c-Met inhibitory effective doses in the low micromolar range while 4-acetamido-4′-isothiocyanatostilbene-2,2′-disulfonic acid (SITS) and 4,4′-dinitrostilbene-2, 2′-disulfonic acid (DNDS) exhibited IC50s 100 to 1000 fold higher. These compounds displayed much greater selectivity for inhibiting c-Met activation compared to similar receptor tyrosine kinases. In addition, DIDS and H2DIDS reduced hepatocyte growth factor (HGF)-induced, but not epidermal growth factor (EGF)-induced, cell scattering, wound healing, and 3-dimensional (3D) proliferation of tumor cell spheroids. In-cell and cell-free assays suggested that DIDS and H2DIDS can inhibit and reverse c-Met phosphorylation, similar to SU11274. Additional data demonstrated that DIDS is tolerable in vivo. These data provide preliminary support for future studies examining DIDS, H2DIDS, and derivatives as potential c-Met therapeutics.
Collapse
Affiliation(s)
- Alana L Gray
- Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA.,Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - David T Coleman
- Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA.,Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - Reneau F Castore
- Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA.,Feist-Weiller Cancer Center, Shreveport, LA, USA
| | | | | | - James A Cardelli
- Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA.,Feist-Weiller Cancer Center, Shreveport, LA, USA
| |
Collapse
|
117
|
Park S, Koh J, Kim DW, Kim M, Keam B, Kim TM, Jeon YK, Chung DH, Heo DS. MET amplification, protein expression, and mutations in pulmonary adenocarcinoma. Lung Cancer 2015; 90:381-7. [DOI: 10.1016/j.lungcan.2015.10.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/20/2015] [Accepted: 10/22/2015] [Indexed: 01/18/2023]
|
118
|
Lu Y, Liu L, Wang Y, Li F, Zhang J, Ye M, Zhao H, Zhang X, Zhang M, Zhao J, Yan B, Yang A, Feng H, Zhang R, Ren X. siRNA delivered by EGFR-specific scFv sensitizes EGFR-TKI-resistant human lung cancer cells. Biomaterials 2015; 76:196-207. [PMID: 26524539 DOI: 10.1016/j.biomaterials.2015.10.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 10/11/2015] [Accepted: 10/14/2015] [Indexed: 12/22/2022]
Abstract
The overexpression of epidermal growth factor receptor (EGFR) is closely associated with a poor outcome in non-small cell lung cancer (NSCLC), and EGFR is an ideal biomarker for the targeted therapy of NSCLC. Although patients with EGFR-activating mutations respond to EGFR tyrosine kinase inhibitors (EGFR-TKIs), they eventually acquire resistance, which typically results from a secondary EGFR mutation or the activation of other signaling pathways. Novel approaches to overcome or prevent EGFR-TKI resistance are clinically important. In this study, we developed an EGFR-scFv-arginine nonamer peptide fusion protein, s-9R, as an siRNA carrier. Here, we show that s-9R effectively and specifically delivers EGFR-siRNAs, KRAS-siRNA and MET-siRNA into NSCLC cells and silences the expression of target genes. The sensitivity of NSCLC cells to gefitinib was restored after treatment with the s-9R/siRNA complex, and the apoptosis rates of the treated cells were significantly higher than those of the control groups. Furthermore, the co-administration of s-9R/siRNA and gefitinib successfully suppressed the progression of H1975 xenograft tumors and extended the life span of tumor-bearing nude mice. Collectively, the results of this study provide not only a new scFv derivative for delivering siRNA into EGFR-overexpressing, TKI-resistant NSCLC cells but also a novel method for overcoming TKI resistance.
Collapse
Affiliation(s)
- Yuan Lu
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Li Liu
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China; Department of Geriatrics, The Central Hospital of Xianyang, 78 Renming East Road, Xianyang 712000, China
| | - Yuan Wang
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Fakai Li
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Jian Zhang
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Hu Zhao
- The Organ Transplant Institute, The Fuzhou General Hospital (DongFang Hospital), Xiamen University, 156 West Ring Road, Fuzhou 350025, China
| | - Xiang Zhang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Mi Zhang
- Department of Respiratory Medicine, Naval General Hospital, 6 Fucheng Road, Beijing 100048, China
| | - Jing Zhao
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Bo Yan
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Angang Yang
- The State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Huasong Feng
- Department of Respiratory Medicine, Naval General Hospital, 6 Fucheng Road, Beijing 100048, China.
| | - Rui Zhang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| | - Xinling Ren
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
119
|
Shen A, Wang L, Huang M, Sun J, Chen Y, Shen YY, Yang X, Wang X, Ding J, Geng M. c-Myc alterations confer therapeutic response and acquired resistance to c-Met inhibitors in MET-addicted cancers. Cancer Res 2015; 75:4548-59. [PMID: 26483207 DOI: 10.1158/0008-5472.can-14-2743] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 07/23/2015] [Indexed: 11/16/2022]
Abstract
Use of kinase inhibitors in cancer therapy leads invariably to acquired resistance stemming from kinase reprogramming. To overcome the dynamic nature of kinase adaptation, we asked whether a signal-integrating downstream effector might exist that provides a more applicable therapeutic target. In this study, we reported that the transcriptional factor c-Myc functions as a downstream effector to dictate the therapeutic response to c-Met inhibitors in c-Met-addicted cancer and derived resistance. Dissociation of c-Myc from c-Met control, likely overtaken by a variety of reprogrammed kinases, led to acquisition of drug resistance. Notably, c-Myc blockade by RNA interference or pharmacologic inhibition circumvented the acquired resistance to c-Met inhibition. Combining c-Myc blockade and c-Met inhibition in MET-amplified patient-derived xenograft mouse models heightened therapeutic activity. Our findings offer a preclinical proof of concept for the application of c-Myc-blocking agents as a tactic to thwart resistance to kinase inhibitors.
Collapse
Affiliation(s)
- Aijun Shen
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai China
| | - Lu Wang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai China
| | - Min Huang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai China
| | - Jingya Sun
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai China
| | - Yi Chen
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai China
| | - Yan-Yan Shen
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai China
| | - Xinying Yang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai China
| | - Xin Wang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai China
| | - Jian Ding
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai China.
| | - Meiyu Geng
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai China.
| |
Collapse
|
120
|
Murat CDB, da Rosa PWL, Fortes MAHZ, Corrêa L, Machado MCC, Novak EM, Siqueira SAC, Pereira MAA, Corrêa-Giannella ML, Giannella-Neto D, Giorgi RR. Differential expression of genes encoding proteins of the HGF/MET system in insulinomas. Diabetol Metab Syndr 2015; 7:84. [PMID: 26435753 PMCID: PMC4591639 DOI: 10.1186/s13098-015-0079-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 09/22/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Insulinomas are the most common functional pancreatic neuroendocrine tumors, whereas histopathological features do not predict their biological behaviour. In an attempt to better understand the molecular processes involved in the tumorigenesis of islet beta cells, the present study evaluated the expression of genes belonging to the hepatocyte growth factor and its receptor (HGF/MET) system, namely, MET, HGF; HGFAC and ST14 (encode HGF activator and matriptase, respectively, two serine proteases that catalyze conversion of pro-HGF to active HGF); and SPINT1 and SPINT2 (encode serine peptidase inhibitors Kunitz type 1 and type 2, respectively, two inhibitors of HGF activator and of matriptase). METHODS Quantitative real-time reverse transcriptase polymerase chain reaction was employed to assess RNA expression of the target genes in 24 sporadic insulinomas: 15 grade 1 (G1), six grade 2 (G2) and three hepatic metastases. Somatic mutations of MET gene were searched by direct sequencing of exons 2, 10, 14, 16, 17 and 19. RESULTS Overexpression of MET was observed in the three hepatic metastases concomitantly with upregulation of the genes encoding HGF and matriptase and downregulation of SPINT1. A positive correlation was observed between MET RNA expression and Ki-67 proliferation index while a negative correlation was detected between SPINT1 expression and the mitotic index. No somatic mutations were found in MET gene. CONCLUSION The final effect of the increased expression of HGF, its activator (matriptase) and its specific receptor (MET) together with a decreased expression of one potent inhibitor of matriptase (SPINT1) is probably a contribution to tumoral progression and metastatization in insulinomas.
Collapse
Affiliation(s)
- Cahuê De Bernardis Murat
- />Laboratório de Endocrinologia Celular e Molecular (LIM-25) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), Av. Dr. Arnaldo, 455, 01246-903 São Paulo, SP Brazil
| | - Paula Waki Lopes da Rosa
- />Laboratório de Endocrinologia Celular e Molecular (LIM-25) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), Av. Dr. Arnaldo, 455, 01246-903 São Paulo, SP Brazil
| | - Maria Angela Henriques Zanella Fortes
- />Laboratório de Endocrinologia Celular e Molecular (LIM-25) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), Av. Dr. Arnaldo, 455, 01246-903 São Paulo, SP Brazil
| | - Luciana Corrêa
- />Departamento de Patologia Oral, Faculdade de Odontologia da Universidade de São Paulo, São Paulo, Brazil
| | | | - Estela Maria Novak
- />Laboratório de Biologia Molecular da Fundação Pró-Sangue Hemocentro de São Paulo, São Paulo, Brazil
| | | | | | - Maria Lucia Corrêa-Giannella
- />Laboratório de Endocrinologia Celular e Molecular (LIM-25) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), Av. Dr. Arnaldo, 455, 01246-903 São Paulo, SP Brazil
- />Centro de Terapia Celular e Molecular (NUCEL/NETCEM) da FMUSP, São Paulo, Brazil
| | - Daniel Giannella-Neto
- />Programa de Pós-Graduação em Medicina, Universidade Nove de Julho—UNINOVE, São Paulo, Brazil
| | - Ricardo Rodrigues Giorgi
- />Laboratório de Endocrinologia Celular e Molecular (LIM-25) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), Av. Dr. Arnaldo, 455, 01246-903 São Paulo, SP Brazil
- />Programa de Pós Graduação em Ciências da Saúde, Universidade de Santo Amaro (UNISA), São Paulo, Brazil
| |
Collapse
|
121
|
Dickgiesser S, Rasche N, Nasu D, Middel S, Hörner S, Avrutina O, Diederichsen U, Kolmar H. Self-Assembled Hybrid Aptamer-Fc Conjugates for Targeted Delivery: A Modular Chemoenzymatic Approach. ACS Chem Biol 2015; 10:2158-65. [PMID: 26131766 DOI: 10.1021/acschembio.5b00315] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Over the past decade, DNA and RNA aptamers have attracted keen research interest due to their ability to specifically bind targets of therapeutic relevance. However, their application is often hampered by a short serum half-life and missing effector functions. Conjugation of aptamers to antibody Fc fragments could improve pharmacokinetics, enable immune effector mechanisms, and provide an option for the introduction of desired payloads (e.g., toxins or fluorescent dyes). We developed a modular scaffold-supported system based on human IgG1 Fc fragments, which allows for its dual functionalization with moieties of interest. In our approach, two bioorthogonal, enzyme-mediated reactions were used in combination with oxime ligation and self-assembly based on PNA-DNA base pairing. Thus, an engineered synthetic peptide nucleic acid (PNA) oligomer was coupled to the C-termini of the Fc dimer upon sequence-specific sortase A-mediated transpeptidation. Hybridization of the resulting Fc-PNA conjugate with a tailored DNA aptamer that binds cancer-related hepatocyte growth factor receptor (c-MET) led to a hybrid construct which showed strong and specific binding to c-MET and was readily internalized by c-MET-overexpressing cells. To install an additional orthogonally addressable site, aldehyde tag technology was applied followed by oxime ligation with an aminooxy-bearing fluorescent dye as model cargo. Delivery of fluorescent probe specifically to c-MET-overexpressing cells was confirmed by flow cytometry. Our approach can provide access to engineered aptamer-Fc conjugates with desired target specificity and cytotoxic payloads.
Collapse
Affiliation(s)
- Stephan Dickgiesser
- Institute
for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Nicolas Rasche
- Protein Engineering
and Antibody Technologies, Merck-Serono, Merck KGaA, Darmstadt, Germany
| | - Daichi Nasu
- Institute
for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Stephen Middel
- Institute
for Organic and Biomolecular Chemistry, Georg-August University Göttingen, Göttingen, Germany
| | - Sebastian Hörner
- Institute
for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Olga Avrutina
- Institute
for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Ulf Diederichsen
- Institute
for Organic and Biomolecular Chemistry, Georg-August University Göttingen, Göttingen, Germany
| | - Harald Kolmar
- Institute
for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| |
Collapse
|
122
|
Sugano T, Seike M, Noro R, Soeno C, Chiba M, Zou F, Nakamichi S, Nishijima N, Matsumoto M, Miyanaga A, Kubota K, Gemma A. Inhibition of ABCB1 Overcomes Cancer Stem Cell-like Properties and Acquired Resistance to MET Inhibitors in Non-Small Cell Lung Cancer. Mol Cancer Ther 2015; 14:2433-40. [PMID: 26351321 DOI: 10.1158/1535-7163.mct-15-0050] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 08/21/2015] [Indexed: 11/16/2022]
Abstract
Patients with non-small cell lung cancer (NSCLC) EGFR mutations have shown a dramatic response to EGFR inhibitors (EGFR-TKI). EGFR T790M mutation and MET amplification have been recognized as major mechanisms of acquired resistance to EGFR-TKI. Therefore, MET inhibitors have recently been used in NSCLC patients in clinical trials. In this study, we tried to identify the mechanism of acquired resistance to MET inhibitors. We analyzed the antitumor effects of two MET inhibitors, PHA-665752 and crizotinib, in 10 NSCLC cell lines. EBC-1 cells with MET amplification were the only cells that were sensitive to both MET inhibitors. We established PHA-665752-resistant EBC-1 cells, namely EBC-1R cells. Activation of KRAS, EGFR, and FGFR2 signaling was observed in EBC-1R cells by FISH and receptor tyrosine kinase phosphorylation antibody arrays. EBC-1R cells also showed overexpression of ATP-binding cassette subfamily B member 1 (ABCB1) as well as phosphorylation of MET. EBC-1R cells grew as cell spheres that exhibited cancer stem cell-like (CSC) properties and epithelial-mesenchymal transition (EMT). The level of miR-138 that targeted ABCB1 was decreased in EBC-1R cells. ABCB1 siRNA and the ABCB1 inhibitor elacridar could reduce sphere numbers and suppress EMT. Elacridar could also reverse resistance to PHA-665752 in EBC-1R cells. Our study demonstrated that ABCB1 overexpression, which was associated with CSC properties and EMT, was involved in the acquired resistance to MET inhibitors. Inhibition of ABCB1 might be a novel therapeutic strategy for NSCLC patients with acquired resistance to MET inhibitors.
Collapse
Affiliation(s)
- Teppei Sugano
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Masahiro Seike
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan.
| | - Rintaro Noro
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Chie Soeno
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Mika Chiba
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Fenfei Zou
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Shinji Nakamichi
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Nobuhiko Nishijima
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Masaru Matsumoto
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Akihiko Miyanaga
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Kaoru Kubota
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Akihiko Gemma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
123
|
Planchard D, Loriot Y, André F, Gobert A, Auger N, Lacroix L, Soria JC. EGFR-independent mechanisms of acquired resistance to AZD9291 in EGFR T790M-positive NSCLC patients. Ann Oncol 2015; 26:2073-8. [PMID: 26269204 DOI: 10.1093/annonc/mdv319] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/20/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND AZD9291 is an oral, irreversible, mutant-selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (EGFR-TKI), which specifically targets both sensitizing and resistant T790M mutations. This compound has shown outstanding activity, in a phase I/II (AURA) trial. However, despite impressive tumor responses in T790M-positive patients, acquired resistance to this drug limits the benefit of this compound. Mutations at the EGFR C797 codon, located within the kinase-binding site, were very recently reported to be a potential mechanism of resistance to AZD9291 in T790M-positive patients. PATIENTS AND METHODS To identify potential mechanisms of resistance to AZD9291, we report here on two patients with resistant biopsy specimens that had been treated with AZD9291. RESULTS We identified in two distinct cases, HER2 and MET amplification by FISH and CGH as a potential mechanism of acquired resistance to third-generation EGFR-TKI. Interestingly, this event occurred with complete loss of the T790M mutation. In one case, we observed a different molecular status at two biopsy sites (the T790M mutation at the primary site and wild-type T790M at the metastatic site with different pathways of acquired resistance to AZD9291). CONCLUSION Our observations suggest that T790M-positive and wild-type T790M clones may coexist at baseline. AZD9291 efficiently suppresses the growth of T790M-positive cells, but a population of wild-type T790M cells at baseline will mediate the development of resistance, here via a by-pass pathway activating either HER2 or MET.
Collapse
Affiliation(s)
- D Planchard
- Department of Medical Oncology, Gustave Roussy, Villejuif
| | - Y Loriot
- Department of Medical Oncology, Gustave Roussy, Villejuif
| | - F André
- Department of Medical Oncology, Gustave Roussy, Villejuif INSERM U981, Gustave Roussy, University Paris XI, Villejuif, France
| | - A Gobert
- Drug Development Department (DITEP), Gustave Roussy, and University Paris-Sud, Villejuif
| | - N Auger
- Department of Medical Biology and Pathology
| | - L Lacroix
- Translational Research Laboratory and BioBank, Gustave Roussy, Villejuif
| | - J C Soria
- Drug Development Department (DITEP), Gustave Roussy, and University Paris-Sud, Villejuif INSERM U981, Gustave Roussy, University Paris XI, Villejuif, France
| |
Collapse
|
124
|
Wei B, Huang QY, Huang SR, Mai W, Zhong XG. MicroRNA‑34a attenuates the proliferation, invasion and metastasis of gastric cancer cells via downregulation of MET. Mol Med Rep 2015; 12:5255-61. [PMID: 26238271 DOI: 10.3892/mmr.2015.4110] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 05/01/2015] [Indexed: 11/06/2022] Open
Abstract
Proliferation, invasion and metastasis are key features of gastric cancer, contributing to high mortality rates in patients with gastric cancer worldwide. As a direct target of p53, the functions of microRNA (miR)‑34a are important, but controversial, in the progression of gastric cancer. In the present study, the clinical importance of miR‑34a in GC specimens (n=40) were investigated and were confirmed in an independent cohort from The Cancer Genome Atlas (TCGA; n=352). The prognostic value of miR‑34a was analyzed using a Kaplan‑Meier survival curve in the TCGA cohort, in combination with complete follow‑up data (n=157). The level of miR‑34a was detected in the human gastric cancer cell line and normal gastric epithelial cell line. The effect of miR‑34a on proliferation and invasion were evaluated using Cell Counting Kit 8, colony formation and cell invasion assays. The molecular basis of miR‑34a was determined by bioinformatics prediction. The correlation between miR‑34a and MET was assessed using reverse transcription‑quantitative polymerase chain reaction and western blot analyses. The results indicated that miR‑34a was downregulated in the gastric cancer tissues, compared with the normal gastric tissues (P<0.01). miR‑34a was negatively correlated with the depth of invasion and lymph node metastasis of gastric cancer (P<0.01). In the TCGA cohort, the levels of miR‑34a were lower in T3 and T4 tumor stages, compared with the level in the T1 stage, and low levels of miR‑34a predicted significantly longer survival rates in patients with GC (P<0.05). miR‑34a also attenuated the proliferation ability, and inhibited the colony formation and cell invasion abilities of the cells (P<0.01). A negative correlation was observed between miR‑34a and MET in gastric cancer (P<0.01; r=‑0.9526), and >60% of cases exhibited consistent expression of miR‑34a and MET in gastric cancer (P<0.01). In conclusion, miR‑34a was associated with the clinicopathological features of gastric cancer and was a valuable predictor of patient prognosis. miR‑34a acted as a tumor suppressor to inhibit gastric cancer proliferation and invasion via the downregulation of MET.
Collapse
Affiliation(s)
- Bin Wei
- Department of Gastroenterology and Peripheral Vascular Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530000, P.R. China
| | - Qiao Ying Huang
- Department of Blood Transfusion, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Shun Rong Huang
- Department of Gastroenterology and Peripheral Vascular Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530000, P.R. China
| | - Wei Mai
- Department of Gastroenterology and Peripheral Vascular Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530000, P.R. China
| | - Xiao Gang Zhong
- Department of Gastroenterology and Peripheral Vascular Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530000, P.R. China
| |
Collapse
|
125
|
Sacco JJ, Clague MJ. Dysregulation of the Met pathway in non-small cell lung cancer: implications for drug targeting and resistance. Transl Lung Cancer Res 2015; 4:242-52. [PMID: 26207212 PMCID: PMC4483475 DOI: 10.3978/j.issn.2218-6751.2015.03.05] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 02/05/2015] [Indexed: 12/16/2022]
Abstract
The receptor tyrosine kinase, Met, orchestrates a complex signalling network that physiologically drives a programme of 'invasive growth'. In cancer however, this process may be co-opted to promote proliferation, survival and metastasis of cancer cells. Met is thus a key therapeutic target, not least in non-small cell lung cancer (NSCLC) where it is one of the most commonly dysregulated driver oncogenes. Identifying robust biomarkers that allow the selection of patients most likely to respond to Met targeted therapies will however be essential to realising their potential. This has been underlined recently by the early termination of three pivotal phase III trials investigating Met targeted agents in NSCLC, all of which failed to show clinical benefit. In contrast to these trials, which were relatively unselective, a couple of early phase trials have recently been instigated that select patients on the basis of Met amplification. While still at an early stage, interim results are relatively encouraging and strengthen the rationale for using Met amplifaction as a biomarker. Here we will discuss this and other aberrations in Met signalling in relation to their significance in the therapeutic targeting of Met.
Collapse
|
126
|
Liu Q, Jin J, Ying J, Sun M, Cui Y, Zhang L, Xu B, Fan Y, Zhang Q. Frequent epigenetic suppression of tumor suppressor gene glutathione peroxidase 3 by promoter hypermethylation and its clinical implication in clear cell renal cell carcinoma. Int J Mol Sci 2015; 16:10636-49. [PMID: 25970749 PMCID: PMC4463666 DOI: 10.3390/ijms160510636] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 04/30/2015] [Accepted: 05/05/2015] [Indexed: 02/07/2023] Open
Abstract
The goal of this study is to identify novel tumor suppressor genes silenced by promoter methylation in clear cell renal cell carcinoma (ccRCC) and discover new epigenetic biomarkers for early cancer detection. Reactive oxygen species (ROS) is a major cause of DNA damage that correlates with cancer initiation and progression. Glutathione peroxidase 3 (GPX3), the only known extracellular glycosylated enzyme of GPXs, is a major scavenger of ROS. GPX3 has been identified as a tumor suppressor in many cancers. However, the role of GPX3 in ccRCC remains unclear. This study aimed to investigate its epigenetic alteration in ccRCC and possible clinicopathological association. In our study, GPX3 methylation and down-regulation were detected in 5 out of 6 ccRCC cell lines and the GPX3 mRNA and protein expression level in ccRCC tumors was significantly lower than in adjacent non-malignant renal tissues (p < 0.0001). Treatment with 5-Aza-2'-deoxycytidine restored GPX3 expression in ccRCC cells. Aberrant methylation was further detected in 77.1% (162/210) of RCC primary tumors, but only 14.6% (7/48) in adjacent non-malignant renal tissues. GPX3 methylation status was significantly associated with higher tumor nuclear grade (p = 0.014). Thus, our results showing frequent GPX3 inactivation by promoter hypermethylation in ccRCC may reveal the failure in the cellular antioxidant system in ccRCC and may be associated with renal tumorigenesis. GPX3 tumor specific methylation may serve as a biomarker for early detection and prognosis prediction of ccRCC.
Collapse
Affiliation(s)
- Qianling Liu
- Department of Urology, Peking University First Hospital and Institute of Urology, National Research Center for Genitourinary Oncology, Beijing 100034, China.
| | - Jie Jin
- Department of Urology, Peking University First Hospital and Institute of Urology, National Research Center for Genitourinary Oncology, Beijing 100034, China.
| | - Jianming Ying
- Department of Pathology, Cancer Institute and Cancer Hospital, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences, Beijing 100021, China.
| | - Mengkui Sun
- Department of Urology, Peking University First Hospital and Institute of Urology, National Research Center for Genitourinary Oncology, Beijing 100034, China.
| | - Yun Cui
- Department of Urology, Peking University First Hospital and Institute of Urology, National Research Center for Genitourinary Oncology, Beijing 100034, China.
| | - Lian Zhang
- Department of Urology, Peking University First Hospital and Institute of Urology, National Research Center for Genitourinary Oncology, Beijing 100034, China.
| | - Ben Xu
- Department of Urology, Peking University First Hospital and Institute of Urology, National Research Center for Genitourinary Oncology, Beijing 100034, China.
| | - Yu Fan
- Department of Urology, Peking University First Hospital and Institute of Urology, National Research Center for Genitourinary Oncology, Beijing 100034, China.
| | - Qian Zhang
- Department of Urology, Peking University First Hospital and Institute of Urology, National Research Center for Genitourinary Oncology, Beijing 100034, China.
| |
Collapse
|
127
|
Pérez-Ramírez C, Cañadas-Garre M, Jiménez-Varo E, Faus-Dáder MJ, Calleja-Hernández MÁ. MET: a new promising biomarker in non-small-cell lung carcinoma. Pharmacogenomics 2015; 16:631-47. [PMID: 25893986 DOI: 10.2217/pgs.15.11] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) leads cancer-related deaths worldwide. Mutations in the kinase domain of the EGFR gene provide sensitivity to tyrosine kinase inhibitors (TKI) drugs. TKI show initial response rates over 75% in mutant EGFR-NSCLC patients, although most of these patients acquire resistance to EGFR inhibitors after therapy. EGFR-TKI resistance mechanisms include amplification in MET and its ligand, and also MET mutations. MET signaling dysregulation has been involved in tumor cell growth, survival, migration and invasion, angiogenesis and activation of several pathways, therefore representing an attractive target for anticancer drug development. In this review, we will discuss MET-related mechanisms of EGFR-TKI resistance in NSCLC, as well as the main drugs targeted to inhibit MET pathway.
Collapse
Affiliation(s)
- Cristina Pérez-Ramírez
- Pharmacogenetics Unit, UGC Provincial de Farmacia de Granada, Instituto de Investigación Biosanitaria de Granada, Complejo Hospitalario Universitario de Granada, Avda Fuerzas Armadas, 2, 18014 Granada, Spain
| | | | | | | | | |
Collapse
|
128
|
Mao ZY, Zhu GQ, Ren L, Guo XC, Su D, Bai L. Prognostic Value of C-met Expression in Cholangiocarcinoma. Technol Cancer Res Treat 2015; 15:227-33. [PMID: 25873560 DOI: 10.1177/1533034615578959] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/19/2015] [Indexed: 12/14/2022] Open
Abstract
Aim: To explore the relationship of clinicopathological features and the proteins of C-met expression in the prognosis of cholangiocarcinoma. Methods: Clinical data and the completed follow-up information of patients with cholangiocarcinoma who underwent cholangiocarcinoma operation from January 2004 to December 2010 were analyzed retrospectively. The relationship of clinicopathological features and C-met in the prognosis of the patients was analyzed. Results: Patients with high expression of C-met had significantly shorter overall survival than those with low expression of C-met, the difference being statistically significant ( P = .003). Patients with high C-met expression had significantly shorter disease-free survival time than those with low expression of C-met, the difference being statistically significant ( P = .009). By COX multivariate analysis, high C-met expression in tumor tissues was an independent risk factor in predicting overall survival and disease-free survival for patients with cholangiocarcinoma ( P = .038, .048, relative risk = 1.390, 1.427). Conclusion: Patients with high C-met expression in cancer tissues had shorter disease-free survival and overall survival. High expression of C-met is an independent risk factor for overall survival and disease-free survival.
Collapse
Affiliation(s)
- Zhi-Yuan Mao
- Department of Oncology, Air Force General Hospital of PLA, Beijing, China
| | - Guang-Qing Zhu
- Department of Oncology, Air Force General Hospital of PLA, Beijing, China
| | - Li Ren
- Department of Pathology, Air Force General Hospital of PLA, Beijing, China
| | - Xiao-Chuan Guo
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Dan Su
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Li Bai
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
129
|
Delaney C, Frank S, Huang RS. Pharmacogenomics of EGFR-targeted therapies in non-small cell lung cancer: EGFR and beyond. CHINESE JOURNAL OF CANCER 2015; 34:149-60. [PMID: 25962919 PMCID: PMC4593375 DOI: 10.1186/s40880-015-0007-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/22/2014] [Indexed: 02/08/2023]
Abstract
Commonly observed aberrations in epidermal growth factor receptor (EGFR) signaling have led to the development of EGFR-targeted therapies for various cancers, including non–small cell lung cancer (NSCLC). EGFR mutations and overexpression have further been shown to modulate sensitivity to these EGFR-targeted therapies in NSCLC and several other types of cancers. However, it is clear that mutations and/or genetic variations in EGFR alone cannot explain all of the variability in the responses of patients with NSCLC to EGFR-targeted therapies. For instance, in addition to EGFR genotype, genetic variations in other members of the signaling pathway downstream of EGFR or variations in parallel receptor tyrosine kinase (RTK) pathways are now recognized to have a significant impact on the efficacy of certain EGFR-targeted therapies. In this review, we highlight the mutations and genetic variations in such genes downstream of EGFR and in parallel RTK pathways. Specifically, the directional effects of these pharmacogenetic factors are discussed with a focus on two commonly prescribed EGFR inhibitors: cetuximab and erlotinib. The results of this comprehensive review can be used to optimize the treatment of NSCLC with EGFR inhibitors. Furthermore, they may provide the rationale for the design of subsequent combination therapies that involve the inhibition of EGFR.
Collapse
Affiliation(s)
- Christopher Delaney
- Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA.
| | - Samuel Frank
- Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA.
| | - R Stephanie Huang
- Department of Medicine, University of Chicago, 900 E 57th street, KCBD room 7148, Chicago, IL, 60637, USA. .,The Affiliated Hospital, School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China.
| |
Collapse
|
130
|
Schmitz K, Koeppen H, Binot E, Fassunke J, Künstlinger H, Ihle MA, Heydt C, Wardelmann E, Büttner R, Merkelbach-Bruse S, Rüschoff J, Schildhaus HU. MET gene copy number alterations and expression of MET and hepatocyte growth factor are potential biomarkers in angiosarcomas and undifferentiated pleomorphic sarcomas. PLoS One 2015; 10:e0120079. [PMID: 25844809 PMCID: PMC4386816 DOI: 10.1371/journal.pone.0120079] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 01/19/2015] [Indexed: 11/19/2022] Open
Abstract
Soft tissue sarcomas are a heterogeneous group of tumors with many different subtypes. In 2014 an estimated 12,020 newly diagnosed cases and 4,740 soft tissue sarcoma related deaths can be expected in the United States. Many soft tissue sarcomas are associated with poor prognosis and therapeutic options are often limited. The evolution of precision medicine has not yet fully reached the clinical treatment of sarcomas since therapeutically tractable genetic changes have not been comprehensively studied so far. We analyzed a total of 484 adult-type malignant mesenchymal tumors by MET fluorescence in situ hybridization and MET and hepatocyte growth factor immunohistochemistry. Eleven different entities were included, among them the most common and clinically relevant subtypes and tumors with specific translocations or complex genetic changes. MET protein expression was observed in 2.6% of the cases, all of which were either undifferentiated pleomorphic sarcomas or angiosarcomas, showing positivity rates of 14% and 17%, respectively. 6% of the tumors showed hepatocyte growth factor overexpression, mainly seen in undifferentiated pleomorphic sarcomas and angiosarcomas, but also in clear cell sarcomas, malignant peripheral nerve sheath tumors, leiomyosarcomas and gastrointestinal stromal tumors. MET and hepatocyte growth factor overexpression were significantly correlated and may suggest an autocrine activation in these tumors. MET FISH amplification and copy number gain were present in 4% of the tumors (15/413). Two samples, both undifferentiated pleomorphic sarcomas, fulfilled the criteria for high level amplification of MET, one undifferentiated pleomorphic sarcoma reached an intermediate level copy number gain, and 12 samples of different subtypes were categorized as low level copy number gains for MET. Our findings indicate that angiosarcomas and undifferentiated pleomorphic sarcomas rather than other frequent adult-type sarcomas should be enrolled in screening programs for clinical trials with MET inhibitors. The screening methods should include both in situ hybridization and immunohistochemistry.
Collapse
Affiliation(s)
- Katja Schmitz
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
- Institute of Pathology, University Hospital Göttingen, Göttingen, Germany
| | - Hartmut Koeppen
- Genentech Inc., South San Francisco, California, United States of America
| | - Elke Binot
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Jana Fassunke
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Helen Künstlinger
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Michaela A. Ihle
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Carina Heydt
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Eva Wardelmann
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | | | - Hans-Ulrich Schildhaus
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
- Institute of Pathology, University Hospital Göttingen, Göttingen, Germany
- * E-mail:
| |
Collapse
|
131
|
Finocchiaro G, Toschi L, Gianoncelli L, Baretti M, Santoro A. Prognostic and predictive value of MET deregulation in non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:83. [PMID: 25992382 PMCID: PMC4416957 DOI: 10.3978/j.issn.2305-5839.2015.03.43] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/11/2015] [Indexed: 12/11/2022]
Abstract
Recent progress in cancer biology has led to the discovery of increasing number of oncogene alterations that have dramatically changed the paradigm of lung cancer treatment. MET is a tyrosine kinase receptor for the hepatocyte growth factor (HGF) that is deregulated in several malignancies, including non-small cell lung cancer (NSCLC). Abnormal MET-HGF signaling pathway activation can occur via different mechanisms, including HGF and/or MET overexpression, MET gene amplification, mutations or rearrangements. MET protein overexpression and increased MET gene number have been identified as poor prognostic factors in several series of surgically resected NSCLC making this receptor an attractive target for cancer treatment. Several clinical trials have recently evaluated the activity of a variety of anti-MET strategies in NSCLC patients with or without molecular selection with a variable degree of success, underscoring the need of establishing the best predictive biomarker for the identification of responding patients.
Collapse
Affiliation(s)
- Giovanna Finocchiaro
- Department of Medical Oncology, Department of Medical Oncology, Istituto Clinico Humanitas IRCCS, Rozzano, Milan, Italy
| | - Luca Toschi
- Department of Medical Oncology, Department of Medical Oncology, Istituto Clinico Humanitas IRCCS, Rozzano, Milan, Italy
| | - Letizia Gianoncelli
- Department of Medical Oncology, Department of Medical Oncology, Istituto Clinico Humanitas IRCCS, Rozzano, Milan, Italy
| | - Marina Baretti
- Department of Medical Oncology, Department of Medical Oncology, Istituto Clinico Humanitas IRCCS, Rozzano, Milan, Italy
| | - Armando Santoro
- Department of Medical Oncology, Department of Medical Oncology, Istituto Clinico Humanitas IRCCS, Rozzano, Milan, Italy
| |
Collapse
|
132
|
Califano R, Morgillo F, De Mello RA, Mountzios G. Role of mesenchymal-epithelial transition amplification in resistance to anti-epidermal growth factor receptor agents. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:81. [PMID: 25992380 PMCID: PMC4416951 DOI: 10.3978/j.issn.2305-5839.2015.03.44] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/06/2015] [Indexed: 11/14/2022]
Abstract
All patients with epidermal growth factor receptor (EGFR) mutant advanced non-small cell lung cancer (NSCLC) treated with an EGFR-tyrosine kinase inhibitor (EGFR-TKI) such as gefitinib, erlotinib or afatinib will progress after a median of 9-12 months. So far, development of a secondary T790M mutation represents the most common (approximately 60%) mechanism of resistance to these drugs. The relative rarity of mesenchymal-epithelial transition (MET) amplification in NSCLC suggests that this event plays a limited role in primary resistance to EGFR-TKI. In contrast, MET gene amplification has been detected as a secondary event representing one of the most relevant mechanisms involved in the acquired resistance to EGFR-TKIs both in preclinical and clinical studies. The aim of this review is to discuss the role of MET amplification as a mechanism of resistance to anti-EGFR therapies and to review strategies which aim at overcoming this mechanism of resistance, including studies assessing drug combinations targeting both EGFR and MET pathways.
Collapse
|
133
|
Zorzetto M, Ferrari S, Saracino L, Inghilleri S, Stella GM. MET genetic lesions in non-small-cell lung cancer: pharmacological and clinical implications. Transl Lung Cancer Res 2015; 1:194-207. [PMID: 25806181 DOI: 10.3978/j.issn.2218-6751.2012.09.03] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 09/14/2012] [Indexed: 12/14/2022]
Abstract
Lung cancer is the leading cause of death for solid tumors worldwide with an annual mortality of over one million. Lung carcinoma includes a series of different diseases which are roughly divided into two groups based on clinical and histo-pathological features: non-small cell lung cancer (NSCLC), accounting for almost 80% of lung cancer diagnosis and small cell lung cancer (SCLC) responsible for the remaining 20%. The NSCLC molecular profile has been deeply investigated; alterations in several oncogenes, tumor suppressor genes and transcription factors have been detected, mainly in adenocarcinomas. Dissection of such a complex scenario represents a still open challenge for both researchers and clinicians. MET, the receptor for Hepatocyte Growth Factor (HGF), has been recently identified as a novel promising target in several human malignancies, including NSCLC. Deregulation of the HGF/MET signaling pathway can occur via different mechanisms, including HGF and/or MET overexpression, MET gene amplification, mutations or rearrangements. While the role of MET mutations in NSCLC is not yet fully understood, MET amplification emerged as a critical event in driving cell survival, with preclinical data suggesting that MET-amplified cell lines are exquisitely sensitive to MET inhibition. True MET amplification, which has been associated with poor prognosis in different retrospective series, is a relatively uncommon event in NSCLC, occurring in 1-7% of unselected cases. Nevertheless, in highly selected cohorts of patients, such as those harboring somatic mutations of EGFR with acquired resistance to EGFR tyrosine kinase inhibitors, MET amplification can be observed in up to 20% of cases. Preclinical data suggested that a treatment approach including a combination of EGFR and MET tyrosine kinases could be an effective strategy in this setting and led to the clinical investigation of multiple MET inhibitors in combination with anti-EGFR agents. Results from ongoing and future trials will clarify the role of anti-MET molecules for the treatment of NSCLC and will provide insights into the most appropriate timing for their use. The present review recapitulates the current knowledge on the role of MET signaling in NSCLC mainly focusing on its implications in molecular diagnostic approach and on the novel targeted inhibitors.
Collapse
Affiliation(s)
- Michele Zorzetto
- Department of Molecular Medicine, - Section of Pneumology, Laboratory of Biochemistry & Genetics; University and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia- Italy
| | - Simona Ferrari
- Department of Molecular Medicine, - Section of Pneumology, Laboratory of Biochemistry & Genetics; University and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia- Italy
| | - Laura Saracino
- Department of Molecular Medicine, - Section of Pneumology, Laboratory of Biochemistry & Genetics; University and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia- Italy
| | - Simona Inghilleri
- Department of Molecular Medicine, - Section of Pneumology, Laboratory of Biochemistry & Genetics; University and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia- Italy
| | - Giulia M Stella
- Department of Molecular Medicine, - Section of Pneumology, Laboratory of Biochemistry & Genetics; University and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia- Italy
| |
Collapse
|
134
|
Menis J, Giaj Levra M, Novello S. MET inhibition in lung cancer. Transl Lung Cancer Res 2015; 2:23-39. [PMID: 25806202 DOI: 10.3978/j.issn.2218-6751.2012.12.04] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 12/17/2012] [Indexed: 01/06/2023]
Abstract
Targeted agents have completely changed cancer treatment strategy, leading it from a "one size fits all" approach to a customized therapy. In this scenario Met, a heterodimere receptor tyrosine kinase deeply involved into embryogenesis and organogenesis, has been introduced many years ago as a potential target for biological agents, becoming "druggable" only in this last period of time. Met can be altered through receptor overexpression, genomic amplification, mutations or alternative splicing, autocrine or paracrine secretion of hepatic growth factor (HGF): these dysregulations stimulate tumorigenesis (in terms of cell-cell detachment, proliferation, invasion, angiogenesis and survival) and metastatization. Met is overexpressed in lung cancer and Met gene amplification can drive the dependency of cell survival and proliferation upon the Met signaling. Both Met overexpression and amplification seem to correlate with poor prognosis. Met amplification is also described to be linked to EGFR acquired resistance. Several Met inhibitors have been tested both in preclinical and human trials, demonstrating activity in lung cancer treatment. This paper aims to summarize data on Met biological function, on its interaction with cell signaling and other pathways and to present data on those Met inhibitors currently under evaluation.
Collapse
Affiliation(s)
- Jessica Menis
- Department of Oncology, A.O.U Santa Maria della Misericordia of Udine, Italy
| | - Matteo Giaj Levra
- Department of Oncology, A.O.U San Luigi - Orbassano, University of Turin, Italy
| | - Silvia Novello
- Department of Oncology, A.O.U San Luigi - Orbassano, University of Turin, Italy
| |
Collapse
|
135
|
Phase I study of the anti-MET antibody onartuzumab in patients with solid tumors and MET-positive lung cancer. Invest New Drugs 2015; 33:632-40. [PMID: 25777467 DOI: 10.1007/s10637-015-0227-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/04/2015] [Indexed: 10/23/2022]
Abstract
Onartuzumab is a monovalent, humanized, monoclonal antibody that showed significant survival benefits in combination with erlotinib in MET-positive non-small-cell lung cancer (NSCLC) in pre-specified subgroup analyses of a randomized phase II study. We conducted a two-stage, open-label, multicenter, phase I study of onartuzumab in Japanese patients. Stage 1 investigated the safety, tolerability, pharmacokinetics (PK), and recommended dose of onartuzumab in patients with solid tumors, and Stage 2 determined the safety, tolerability, and PK of onartuzumab plus erlotinib in patients with MET-positive NSCLC. Nine patients received onartuzumab monotherapy (4, 15, or 30 mg/kg on Day 1 of each 21-day cycle) in Stage 1, and six patients received onartuzumab (15 mg/kg) plus erlotinib (150 mg/day) in Stage 2. There were no dose-limiting toxicities in either stage. Serious adverse events (AEs) occurred in one patient in Stage 1 (convulsion), and two patients in Stage 2 (once case each of diarrhea, vomiting, and pulmonary embolism), but there were no grade 4 AEs or AEs leading to death. Onartuzumab PKs were linear in the dose range of 4 to 30 mg/kg, and were not affected by co-administration with erlotinib. PK parameters of onartuzumab were similar to those reported in non-Japanese patients. A partial response was observed in a patient with MET immunohistochemistry 3+ NSCLC without MET gene amplification. Based on these results, the recommended dose of onartuzumab in Japanese patients with solid tumors is 15 mg/kg every 21 days. The combination of onartuzumab with erlotinib is feasible in Japanese patients with MET-positive lung cancer.
Collapse
|
136
|
Yang X, Yang Y, Tang S, Tang H, Yang G, Xu Q, Wu J. Anti-tumor effect of polysaccharides from Scutellaria barbata D. Don on the 95-D xenograft model via inhibition of the C-met pathway. J Pharmacol Sci 2015; 125:255-63. [PMID: 25048016 DOI: 10.1254/jphs.13276fp] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Polysaccharides isolated from Scutellaria barbata (PSB) have been reported to have anti-tumor effects. To investigate the underlying mechanism, a highly invasive, metastatic and phospho-c-Met overexpression lung carcinoma cell, 95-D cell line was used. The results showed that in vitro, PSB not only could inhibit the proliferation of 95-D cell line (IC(50) = 35.2 μg/mL), but also down-regulated the expression of phospho-c-Met and its downstream signaling molecules including phospho-Erk and phospho-Akt. In vivo, PSB inhibited tumor growth in the 95-D subcutaneous xenograft model in a dose-dependent manner; after once-daily intraperitoneal injection for 3 weeks, tumor growth inhibition T/C ratio for 100 and 200 mg/kg treatments was 42.72% and 13.6%, respectively. In the end of the in vivo study, tumor tissues were harvested for further evaluation of the phosphorylation level of c-Met, AKT, and ERK. Ex vivo results demonstrated that the phosphorylation of c-Met and its downstream signaling molecules were also significantly inhibited by PSB. Immunohistochemistry analysis showed dose-dependent inhibition of tumor cell proliferation (Ki67) and reduction of microvessel density (CD31). In summary, the results indicated that PSB exerted anti-tumor growth activity on human lung cancer 95-D in vitro and in vivo by directly regulating the c-Met signaling pathway and the anti-tumor effects were mainly based on its anti-proliferation and anti-angiogenesis action.
Collapse
Affiliation(s)
- Xiaokun Yang
- Department of Dermatology, Daping Hospital, Third Military Medical University, China
| | | | | | | | | | | | | |
Collapse
|
137
|
Hirsh V. The treatment of metastatic non-small cell lung cancer in a new era of personalized medicine. Front Oncol 2015; 5:20. [PMID: 25692098 PMCID: PMC4315106 DOI: 10.3389/fonc.2015.00020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/16/2015] [Indexed: 01/28/2023] Open
Affiliation(s)
- Vera Hirsh
- Department of Medical Oncology, McGill University Health Centre , Montreal, QC , Canada
| |
Collapse
|
138
|
Calles A, Kwiatkowski N, Cammarata BK, Ercan D, Gray NS, Jänne PA. Tivantinib (ARQ 197) efficacy is independent of MET inhibition in non-small-cell lung cancer cell lines. Mol Oncol 2015; 9:260-9. [PMID: 25226813 PMCID: PMC5528687 DOI: 10.1016/j.molonc.2014.08.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 08/21/2014] [Indexed: 01/15/2023] Open
Abstract
MET targeted therapies are under clinical evaluation for non-small-cell lung cancer (NSCLC) patients. Tyrosine kinase inhibitors (TKI) against MET have varying degrees of specificity. Tivantinib (ARQ 197) is reported to be a non-ATP competitive selective MET inhibitor. We aimed to compare the activity of tivantinib to established MET TKIs in a panel of NSCLC cell lines characterized by their MET dependency and by different relevant genotypes. A549, H3122, PC9 and HCC827, their respective resistant clones PC9 GR4 and HCC827 GR6 and the MET amplified cell lines H1993 and EBC-1 were treated in vitro with tivantinib, crizotinib or PHA-665752. Crizotinib and PHA-665752 showed growth inhibition restricted to MET dependent cell lines. The pattern of activity was related to MET inhibition and downstream signaling inhibition of AKT and ERK1/2, resulting in G0/G1 cycle arrest and apoptosis. In contrast, tivantinib possessed more potent anti-proliferative activity that was not restricted to only MET dependent cell lines. Tivantinib did not inhibit cellular MET activity or phosphorylation of downstream signaling proteins AKT or ERK1/2 in either MET dependent or independent cell lines. Cell cycle analysis demonstrated that tivantinib induced a G2/M arrest and induced apoptosis. Tivantinib but not crizotinib effected microtubule dynamics, disrupting mitotic spindles by a mechanism consistent with it functioning as a microtubule depolymerizer. Tivantinib activity is independent of MET signaling in NSCLC and suggests alternative mechanisms of action that should be considered when interpreting the results from on-going clinical studies.
Collapse
Affiliation(s)
- Antonio Calles
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nicholas Kwiatkowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Bernard K Cammarata
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Dalia Ercan
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
139
|
Morgensztern D, Campo MJ, Dahlberg SE, Doebele RC, Garon E, Gerber DE, Goldberg SB, Hammerman PS, Heist R, Hensing T, Horn L, Ramalingam SS, Rudin CM, Salgia R, Sequist L, Shaw AT, Simon GR, Somaiah N, Spigel DR, Wrangle J, Johnson D, Herbst RS, Bunn P, Govindan R. Molecularly targeted therapies in non-small-cell lung cancer annual update 2014. J Thorac Oncol 2015; 10:S1-63. [PMID: 25535693 PMCID: PMC4346098 DOI: 10.1097/jto.0000000000000405] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There have been significant advances in the understanding of the biology and treatment of non-small-cell lung cancer (NSCLC) during the past few years. A number of molecularly targeted agents are in the clinic or in development for patients with advanced NSCLC. We are beginning to understand the mechanisms of acquired resistance after exposure to tyrosine kinase inhibitors in patients with oncogene addicted NSCLC. The advent of next-generation sequencing has enabled to study comprehensively genomic alterations in lung cancer. Finally, early results from immune checkpoint inhibitors are very encouraging. This review summarizes recent advances in the area of cancer genomics, targeted therapies, and immunotherapy.
Collapse
Affiliation(s)
- Daniel Morgensztern
- Department of Medical Oncology, Washington University School of Medicine, Saint Louis, MO
| | - Meghan J. Campo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Suzanne E. Dahlberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston MA
| | - Robert C. Doebele
- Department of Medical Oncology, University of Colorado School of Medicine and University of Colorado Cancer Center, Aurora, CO
| | - Edward Garon
- UCLA Santa Monica Hematology Oncology, Santa Monica, CA
| | - David E. Gerber
- Division of Hematology-Oncology, Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Sarah B. Goldberg
- Department of Medical Oncology, Yale School of Medicine and Cancer Center, New Haven, CT
| | | | - Rebecca Heist
- Department of Oncology, Massachusetts General Hospital, Boston, MA
| | - Thomas Hensing
- Department of Oncology, The University of Chicago Medicine, Chicago, IL
| | - Leora Horn
- Division of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Suresh S. Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA
| | | | - Ravi Salgia
- Department of Oncology, The University of Chicago Medicine, Chicago, IL
| | - Lecia Sequist
- Department of Oncology, Massachusetts General Hospital, Boston, MA
| | - Alice T. Shaw
- Department of Oncology, Massachusetts General Hospital, Boston, MA
| | - George R. Simon
- Division of Hematology-Oncology, Medical University of South Carolina, Charleston, SC
| | - Neeta Somaiah
- Division of Hematology-Oncology, Medical University of South Carolina, Charleston, SC
| | | | - John Wrangle
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - David Johnson
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Roy S. Herbst
- Department of Medical Oncology, Yale School of Medicine and Cancer Center, New Haven, CT
| | - Paul Bunn
- Division of Medical Oncology, University of Colorado Denver School of Medicine, Denver, CO
| | - Ramaswamy Govindan
- Department of Medical Oncology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
140
|
Abstract
INTRODUCTION The mesenchymal-epithelial transition (MET) protein is the only known receptor for hepatocyte growth factor and has recently been identified as a novel promising target in several human cancers, including NSCLC. Activation of the MET signaling pathway can occur via different mechanisms. A number of compounds targeting MET have been evaluated in clinical trials, and recent clinical data have begun to afford some insight into the tumor types and patient populations that might benefit from treatment with MET pathway inhibitors. AREAS COVERED We review recent publications and information disclosed at public conferences and summarize the epidemiology of dysregulation of MET signaling, and the associations thereof with other driver genes and therapeutic inhibitors useful to treat NSCLC. EXPERT OPINION The MET pathway is emerging as a target for advanced NSCLC that is either resistant to EGFR tyrosine kinase inhibitors or that arises de novo. MET inhibitors currently being evaluated in clinical trials have yielded compelling evidence of clinical activities when used to treat various solid tumors, especially NSCLC. Remaining challenges are the identification of patient populations who might benefit from the use of MET inhibitors, and the most effective diagnostic methods for such patients.
Collapse
Affiliation(s)
- Anna Li
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Science , 106 Zhongshan 2nd Road, Guangzhou 510080 , PR China +86 20 83877855 ; +86 20 83827712 ;
| | | | | |
Collapse
|
141
|
Schildhaus HU, Schultheis AM, Rüschoff J, Binot E, Merkelbach-Bruse S, Fassunke J, Schulte W, Ko YD, Schlesinger A, Bos M, Gardizi M, Engel-Riedel W, Brockmann M, Serke M, Gerigk U, Hekmat K, Frank KF, Reiser M, Schulz H, Krüger S, Stoelben E, Zander T, Wolf J, Buettner R. MET amplification status in therapy-naïve adeno- and squamous cell carcinomas of the lung. Clin Cancer Res 2014; 21:907-15. [PMID: 25492085 DOI: 10.1158/1078-0432.ccr-14-0450] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE MET is a potential therapeutic target in lung cancer and both MET tyrosine kinase inhibitors and monoclonal antibodies have entered clinical trials. MET signaling can be activated by various mechanisms, including gene amplification. In this study, we aimed to investigate MET amplification status in adeno- and squamous cell carcinomas of the lung. We propose clearly defined amplification scores and provide epidemiologic data on MET amplification in lung cancer. EXPERIMENTAL DESIGN We evaluated the prevalence of increased MET gene copy numbers in 693 treatment-naïve cancers by FISH, defined clear cutoff criteria, and correlated FISH results to MET IHC. RESULTS Two thirds (67%) of lung cancers do not have gains in MET gene copy numbers, whereas 3% show a clear-cut high-level amplification (MET/centromer7 ratio ≥2.0 or average gene copy number per nucleus ≥6.0 or ≥10% of tumor cells containing ≥15 MET copies). The remaining cases can be subdivided into intermediate- (6%) and low-level gains (24%). Importantly, MET amplifications occur at equal frequencies in squamous and adenocarcinomas without or with EGFR or KRAS mutations. CONCLUSION MET amplification is not a mutually exclusive genetic event in therapy-naïve non-small cell lung cancer. Our data suggest that it might be useful to determine MET amplification (i) before EGFR inhibitor treatment to identify possible primary resistance to anti-EGFR treatment, and (ii) to select cases that harbor KRAS mutations additionally to MET amplification and, thus, may not benefit from MET inhibition. Furthermore, our study provides comprehensive epidemiologic data for upcoming trials with various MET inhibitors.
Collapse
Affiliation(s)
- Hans-Ulrich Schildhaus
- Institute of Pathology, University Hospital Cologne, Cologne, Germany. Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany. Institute of Pathology, University Hospital Göttingen, Göttingen, Germany.
| | - Anne M Schultheis
- Institute of Pathology, University Hospital Cologne, Cologne, Germany. Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Josef Rüschoff
- Institute of Pathology Nordhessen, Kassel, Germany. Targos Molecular Pathology, Kassel, Germany
| | - Elke Binot
- Institute of Pathology, University Hospital Cologne, Cologne, Germany. Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Institute of Pathology, University Hospital Cologne, Cologne, Germany. Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Jana Fassunke
- Institute of Pathology, University Hospital Cologne, Cologne, Germany. Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Wolfgang Schulte
- Thoraxzentrum im Malteser Krankenhaus Bonn/Rhein-Sieg, Bonn, Germany
| | - Yon-Dschun Ko
- Johanniter-Krankenhaus, Evangelische Kliniken Bonn GmbH, Bonn, Germany
| | | | - Marc Bos
- Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany. Department I of Internal Medicine and Centre of Integrated Oncology Köln-Bonn, University of Cologne, Cologne, Germany
| | - Masyar Gardizi
- Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany. Department I of Internal Medicine and Centre of Integrated Oncology Köln-Bonn, University of Cologne, Cologne, Germany
| | | | - Michael Brockmann
- Institute of Pathology, Kliniken der Stadt Köln GmbH, Cologne, Germany
| | | | - Ulrich Gerigk
- Thoraxzentrum im Malteser Krankenhaus Bonn/Rhein-Sieg, Bonn, Germany
| | - Khosro Hekmat
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Konrad F Frank
- Department III of Internal Medicine, University of Cologne, Cologne, Germany
| | - Marcel Reiser
- PIOH-Praxis Internistischer Onkologie und Hämatologie Köln, Cologne, Germany
| | - Holger Schulz
- PIOH-Praxis Internistischer Onkologie und Hämatologie Frechen, Frechen, Germany
| | - Stefan Krüger
- Florence Nightingale Krankenhaus Düsseldorf Kaiserswerth, Düsseldorf, Germany
| | - Erich Stoelben
- Lungenklinik Merheim, Kliniken der Stadt Köln GmbH, Cologne, Germany
| | - Thomas Zander
- Department I of Internal Medicine and Centre of Integrated Oncology Köln-Bonn, University of Cologne, Cologne, Germany. NGM, Network Genomic Medicine and GCGC, Gastrointestinal Cancer Group Cologne, Germany
| | - Jürgen Wolf
- Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany. Department I of Internal Medicine and Centre of Integrated Oncology Köln-Bonn, University of Cologne, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany. Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany. Targos Molecular Pathology, Kassel, Germany
| |
Collapse
|
142
|
Abstract
Hepatitis C virus (HCV) is one of the major etiologic agents of liver cancer. HCV is an RNA virus that, unlike hepatitis B virus, is unable to integrate into the host genome. Through complex interactions between viral and host proteins that induce host responses and promote inflammation, fibrosis, and ultimately cirrhosis, HCV infection can result in the development of hepatocellular carcinoma (HCC). The HCV oncogenic process involves genetic and epigenetic alterations and oncogenic effects mediated by viral proteins in the activation of cellular oncogenes, inactivation of tumor-suppressor genes, and dysregulation of multiple signal-transduction pathways. Advances in genetics and gene expression profiling have enhanced our current understanding of the pathways involved in HCV-associated liver cancer development. In this review, we summarize the current understanding of mechanisms of hepatocarcinogenesis induced by HCV infection.
Collapse
Affiliation(s)
- Ming V Lin
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114; , ,
| | | | | |
Collapse
|
143
|
Agwa ES, Ma PC. Targeting the MET receptor tyrosine kinase in non-small cell lung cancer: emerging role of tivantinib. Cancer Manag Res 2014; 6:397-404. [PMID: 25328417 PMCID: PMC4198276 DOI: 10.2147/cmar.s37345] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
MET receptor tyrosine kinase and its natural ligand, hepatocyte growth factor, have been implicated in a variety of cancers, including non-small cell lung cancer (NSCLC). Mechanisms by which cellular deregulation of MET occurs include overexpression, genomic amplification, mutation, or alternative splicing. MET overexpression or activation is a known cause of acquired resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors in NSCLC. Inhibition of MET signaling in these EGFR tyrosine kinase inhibitor-resistant cells may potentially restore sensitivity to EGFR inhibitors. Tivantinib (ARQ 197), reported as a small-molecule MET inhibitor, has demonstrated antitumor activity in early clinical studies. This review focuses on MET and lung cancer, the clinical development of tivantinib, the clinical trials of tivantinib in NSCLC to date, its current/emerging role in the management of NSCLC, and future directions.
Collapse
Affiliation(s)
- Eberechi S Agwa
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Patrick C Ma
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
144
|
Liu L, Zeng W, Wortinger MA, Yan SB, Cornwell P, Peek VL, Stephens JR, Tetreault JW, Xia J, Manro JR, Credille KM, Ballard DW, Brown-Augsburger P, Wacheck V, Chow CK, Huang L, Wang Y, Denning I, Davies J, Tang Y, Vaillancourt P, Lu J. LY2875358, a neutralizing and internalizing anti-MET bivalent antibody, inhibits HGF-dependent and HGF-independent MET activation and tumor growth. Clin Cancer Res 2014; 20:6059-70. [PMID: 25231402 DOI: 10.1158/1078-0432.ccr-14-0543] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE MET, the receptor for hepatocyte growth factor (HGF), has been implicated in driving tumor proliferation and metastasis. High MET expression is correlated with poor prognosis in multiple cancers. Activation of MET can be induced either by HGF-independent mechanisms such as gene amplification, specific genetic mutations, and transcriptional upregulation or by HGF-dependent autocrine or paracrine mechanisms. EXPERIMENTAL DESIGN/RESULTS Here, we report on LY2875358, a novel humanized bivalent anti-MET antibody that has high neutralization and internalization activities, resulting in inhibition of both HGF-dependent and HGF-independent MET pathway activation and tumor growth. In contrast to other bivalent MET antibodies, LY2875358 exhibits no functional agonist activity and does not stimulate biologic activities such as cell proliferation, scattering, invasion, tubulogenesis, or apoptosis protection in various HGF-responsive cells and no evidence of inducing proliferation in vivo in a monkey toxicity study. LY2875358 blocks HGF binding to MET and HGF-induced MET phosphorylation and cell proliferation. In contrast to the humanized one-armed 5D5 anti-MET antibody, LY2875358 induces internalization and degradation of MET that inhibits cell proliferation and tumor growth in models where MET is constitutively activated. Moreover, LY2875358 has potent antitumor activity in both HGF-dependent and HGF-independent (MET-amplified) xenograft tumor models. Together, these findings indicate that the mechanism of action of LY2875358 is different from that of the one-armed MET antibody. CONCLUSIONS LY2875358 may provide a promising therapeutic strategy for patients whose tumors are driven by both HGF-dependent and HGF-independent MET activation. LY2875358 is currently being investigated in multiple clinical studies.
Collapse
Affiliation(s)
- Ling Liu
- Biotechnology Discovery Research,
| | - Wei Zeng
- Biotechnology Discovery Research
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yong Wang
- Discovery Chemistry Research and Technologies. Lilly Research Laboratories, A Division of Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana
| | | | | | | | | | | |
Collapse
|
145
|
Pennacchietti S, Cazzanti M, Bertotti A, Rideout WM, Han M, Gyuris J, Perera T, Comoglio PM, Trusolino L, Michieli P. Microenvironment-Derived HGF Overcomes Genetically Determined Sensitivity to Anti-MET Drugs. Cancer Res 2014; 74:6598-609. [DOI: 10.1158/0008-5472.can-14-0761] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
146
|
Abstract
Cancers arise through the progression of multiple genetic and epigenetic defects that lead to deregulation of numerous signalling networks. However, the last decade has seen the development of the concept of 'oncogene addiction', where tumours appear to depend on a single oncogene for survival. RNAi has provided an invaluable tool in the identification of these oncogenes and oncogene-dependent cancers, and also presents great potential as a novel therapeutic strategy against them. Although RNAi therapeutics have demonstrated effective killing of oncogene-dependent cancers in vitro, their efficacy in vivo is severely limited by effective delivery systems. Several virus-based RNAi delivery strategies have been explored, but problems arose associated with high immunogenicity, random genome integration and non-specific targeting. This has directed efforts towards non-viral formulations, including delivery systems based on virus-like particles, liposomes and cationic polymers, which can circumvent some of these problems by immunomasking and the use of specific tumour-targeting ligands. This review outlines the prevalence of oncogene-dependent cancers, evaluates the potential of RNAi-based therapeutics and assesses the relative strengths and weaknesses of different approaches to targeted RNAi delivery.
Collapse
|
147
|
Zhao W, Damodaran S, Villalona-Calero MA. From single-gene to multiplex analysis in lung cancer, challenges and accomplishments: a review of a single institution's experience. Lung Cancer Manag 2014; 4:315-324. [PMID: 25580159 DOI: 10.2217/lmt.14.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Molecular selection has led to the successful use of novel tyrosine kinase inhibitors (TKIs) in non-small-cell lung cancers (NSCLCs). For instance, mutations in EGFR and translocations and fusions in ALK render tumor cells sensitive to some TKIs, leading to substantial clinical benefits. Molecular testing such as DNA sequencing or fragment analysis following PCR, and evaluation of copy number and gene positioning by FISH, have been developed and used clinically to identify mutations/fusions. Meanwhile, TKIs to target actionable mutations/fusions in several other oncogenes are being evaluated. High-throughput sequencing can provide therapy-predictive information as well as identify novel targetable genomic alterations. In this article, we report our experience enabling single-gene testing, and our evolution to panel-based next-generation sequencing.
Collapse
Affiliation(s)
- Weiqiang Zhao
- Departments of Internal Medicine & Pathology, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA ; Comprehensive Cancer Center, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA
| | - Senthilkumar Damodaran
- Departments of Internal Medicine & Pathology, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA ; Comprehensive Cancer Center, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA
| | - Miguel A Villalona-Calero
- Departments of Internal Medicine & Pathology, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA ; Comprehensive Cancer Center, The Ohio State University, 320 W, 10th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
148
|
Mathur A, Ware C, Davis L, Gazdar A, Pan BS, Lutterbach B. FGFR2 is amplified in the NCI-H716 colorectal cancer cell line and is required for growth and survival. PLoS One 2014; 9:e98515. [PMID: 24968263 PMCID: PMC4072591 DOI: 10.1371/journal.pone.0098515] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/02/2014] [Indexed: 12/21/2022] Open
Abstract
Aberrant kinase activation resulting from mutation, amplification, or translocation can drive growth and survival in a subset of human cancer. FGFR2 is amplified in breast and gastric cancer, and we report here the first characterization of FGFR2 gene amplification in colorectal cancer in the NCI-H716 colorectal cancer cell line. FGFR2 is highly expressed and activated in NCI-H716 cells, and FGFR selective small molecule inhibitors or FGFR2 shRNA strongly inhibited cell viability in vitro, indicating “addiction” of NCI-H716 cells to FGFR2. NCI-H716 growth in a xenograft model was also inhibited by an FGFR small molecule inhibitor. FGFR2 was required for activation of multiple downstream signaling proteins including AKT, ERK, S6RP and NFKB. Inhibition of downstream kinases such as AKT or ERK alone had modest effects on proliferation, whereas combined inhibition of AKT and ERK signaling resulted in a loss of viability similar to FGFR2 inhibition. We identified elevated FGFR2 expression in a small subset of primary colorectal cancer, however FGFR2 amplification was not observed. Although FGFR2 amplification is not common in primary colon cancer or lymph node and liver metastases, other subsets of colorectal cancer such as ascites, from which the NCI-H716 cell line was derived, have yet to be tested. These results suggest that emerging FGFR inhibitor therapeutics may have efficacy in a subset of colon cancer driven by FGFR2 amplification.
Collapse
Affiliation(s)
- Anjili Mathur
- Merck Research Labs, Boston, Massachusetts, United States of America
| | - Christopher Ware
- Merck Research Labs, Boston, Massachusetts, United States of America
| | - Lenora Davis
- Merck Research Labs, Boston, Massachusetts, United States of America
| | - Adi Gazdar
- Hamon Center for Therapeutic Oncology, University of Texas Southwestern, Dallas, Texas, United States of America
| | - Bo-Sheng Pan
- Merck Research Labs, Boston, Massachusetts, United States of America
| | - Bart Lutterbach
- Merck Research Labs, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
149
|
Cooper WA, Lam DCL, O'Toole SA, Minna JD. Molecular biology of lung cancer. J Thorac Dis 2014; 5 Suppl 5:S479-90. [PMID: 24163741 DOI: 10.3978/j.issn.2072-1439.2013.08.03] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 08/01/2013] [Indexed: 12/16/2022]
Abstract
Lung cancers are characterised by abundant genetic diversity with relatively few recurrent mutations occurring at high frequency. However, the genetic alterations often affect a common group of oncogenic signalling pathways. There have been vast improvements in our understanding of the molecular biology that underpins lung cancer in recent years and this has led to a revolution in the diagnosis and treatment of lung adenocarcinomas (ADC) based on the genotype of an individual's tumour. New technologies are identifying key and potentially targetable genetic aberrations not only in adenocarcinoma but also in squamous cell carcinoma (SCC) of the lung. Lung cancer mutations have been identified in v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS), epidermal growth factor receptor (EGFR), BRAF and the parallel phosphatidylinositol 3-kinase (PI3K) pathway oncogenes and more recently in MEK and HER2 while structural rearrangements in ALK, ROS1 and possibly rearranged during transfection (RET) provide new therapeutic targets. Amplification is another mechanism of activation of oncogenes such as MET in adenocarcinoma, fibroblastgrowth factor receptor 1 (FGFR1) and discoidin domain receptor 2 (DDR2) in SCC. Intriguingly, many of these genetic alternations are associated with smoking status and with particular racial and gender differences, which may provide insight into the mechanisms of carcinogenesis and role of host factors in lung cancer development and progression. The role of tumour suppressor genes is increasingly recognised with aberrations reported in TP53, PTEN, RB1, LKB11 and p16/CDKN2A. Identification of biologically significant genetic alterations in lung cancer that lead to activation of oncogenes and inactivation of tumour suppressor genes has the potential to provide further therapeutic opportunities. It is hoped that these discoveries may make a major contribution to improving outcome for patients with this poor prognosis disease.
Collapse
Affiliation(s)
- Wendy A Cooper
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, Sydney, NSW, Australia; ; School of Medicine, University of Western Sydney, NSW, Australia
| | | | | | | |
Collapse
|
150
|
Martin V, Corso S, Comoglio PM, Giordano S. Increase of MET gene copy number confers resistance to a monovalent MET antibody and establishes drug dependence. Mol Oncol 2014; 8:1561-74. [PMID: 25011627 DOI: 10.1016/j.molonc.2014.06.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 06/16/2014] [Accepted: 06/16/2014] [Indexed: 12/14/2022] Open
Abstract
The relevant role in cancer played by the tyrosine kinase receptor encoded by the MET oncogene led to the development of specific inhibitors, some of which are now in advanced phases of clinical trials. Previous experience has shown that the main limit to the efficacy of most targeted treatments is the advent of resistance. Mechanisms underlying resistance to MET-specific small tyrosine kinase inhibitors (TKIs) have been already described, while nothing is known about resistance to MET monoclonal antibodies, nor about bypassing resistance to chemical TKIs by antibodies or vice-versa. EBC1 lung cancer cells are MET-addicted as a consequence of gene amplification and thus sensitive to MET inhibitors, including the monovalent form of a MET monoclonal antibody (MV-DN30). We generated cells resistant to this antibody and found that resistance was due to a further increase of gene copy number and a dramatic overexpression of the MET receptor. Such an excess of expression saturated the 'shedding' activity of MV-DN30, and prevented both the efficient down-regulation of the MET receptor from the surface and the inhibition of the ensuing constitutive activation. Notably, antibody-resistant cells remained MET-'addicted' and were still sensitive to MET TKIs. Moreover, antibody-resistant cells became 'drug-dependent', since the removal of MV-DN30 led them to death due to excess of signal. In the mirror experiment, cells made resistant to MET-specific TKIs were still sensitive to treatment with the antibody MV-DN30. These findings suggest that a discontinuous, combined treatment by antibodies and chemical kinase inhibitors may increase the clinical response and bypass resistance to anti-MET targeted therapies.
Collapse
Affiliation(s)
- Valentina Martin
- University of Torino, Department of Oncology, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy; Candiolo Cancer Institute-FPO, IRCCS, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy
| | - Simona Corso
- University of Torino, Department of Oncology, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy; Candiolo Cancer Institute-FPO, IRCCS, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy.
| | - Paolo M Comoglio
- University of Torino, Department of Oncology, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy; Candiolo Cancer Institute-FPO, IRCCS, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy
| | - Silvia Giordano
- University of Torino, Department of Oncology, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy; Candiolo Cancer Institute-FPO, IRCCS, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy.
| |
Collapse
|