101
|
Yang OC, Maxwell PH, Pollard PJ. Renal cell carcinoma: translational aspects of metabolism and therapeutic consequences. Kidney Int 2013; 84:667-81. [DOI: 10.1038/ki.2013.245] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 03/12/2013] [Accepted: 03/14/2013] [Indexed: 02/08/2023]
|
102
|
Jacobson O, Chen X. Interrogating tumor metabolism and tumor microenvironments using molecular positron emission tomography imaging. Theranostic approaches to improve therapeutics. Pharmacol Rev 2013; 65:1214-56. [PMID: 24064460 PMCID: PMC3799232 DOI: 10.1124/pr.113.007625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Positron emission tomography (PET) is a noninvasive molecular imaging technology that is becoming increasingly important for the measurement of physiologic, biochemical, and pharmacological functions at cellular and molecular levels in patients with cancer. Formation, development, and aggressiveness of tumor involve a number of molecular pathways, including intrinsic tumor cell mutations and extrinsic interaction between tumor cells and the microenvironment. Currently, evaluation of these processes is mainly through biopsy, which is invasive and limited to the site of biopsy. Ongoing research on specific target molecules of the tumor and its microenvironment for PET imaging is showing great potential. To date, the use of PET for diagnosing local recurrence and metastatic sites of various cancers and evaluation of treatment response is mainly based on [(18)F]fluorodeoxyglucose ([(18)F]FDG), which measures glucose metabolism. However, [(18)F]FDG is not a target-specific PET tracer and does not give enough insight into tumor biology and/or its vulnerability to potential treatments. Hence, there is an increasing need for the development of selective biologic radiotracers that will yield specific biochemical information and allow for noninvasive molecular imaging. The possibility of cancer-associated targets for imaging will provide the opportunity to use PET for diagnosis and therapy response monitoring (theranostics) and thus personalized medicine. This article will focus on the review of non-[(18)F]FDG PET tracers for specific tumor biology processes and their preclinical and clinical applications.
Collapse
Affiliation(s)
- Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD.
| | | |
Collapse
|
103
|
Abstract
In a relatively short period of time, monoclonal antibodies have entered the mainstream of cancer therapy. Their first use was as antagonists of oncogenic receptor tyrosine kinases, but today monoclonal antibodies have emerged as long-sought vehicles for the targeted delivery of potent chemotherapeutic agents and as powerful tools to manipulate anticancer immune responses. With ever more promising results from the clinic, the future will likely see continued growth in the discovery and development of therapeutic antibodies and their derivatives.
Collapse
Affiliation(s)
- Mark X Sliwkowski
- Genentech, Incorporated, 1 DNA Way, South San Francisco, CA 94080, USA.
| | | |
Collapse
|
104
|
Slørdahl TS, Denayer T, Moen SH, Standal T, Børset M, Ververken C, Rø TB. Anti-c-MET Nanobody - a new potential drug in multiple myeloma treatment. Eur J Haematol 2013; 91:399-410. [PMID: 23952536 DOI: 10.1111/ejh.12185] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND c-MET is the tyrosine kinase receptor of the hepatocyte growth factor (HGF). HGF-c-MET signaling is involved in many human malignancies, including multiple myeloma (MM). Recently, multiple agents have been developed directed to interfere at different levels in HGF-c-MET signaling pathway. Nanobodies are therapeutic proteins based on the smallest functional fragments of heavy-chain-only antibodies. In this study, we wanted to determine the anticancer effect of a novel anti-c-MET Nanobody in MM. METHODS We examined the effects of an anti-c-MET Nanobody on thymidine incorporation, migration, adhesion of MM cells, and osteoblastogenesis in vitro. Furthermore, we investigated the effects of the Nanobody on HGF-dependent c-MET signaling by Western blotting. RESULTS We show that the anti-c-MET Nanobody effectively inhibited thymidine incorporation of ANBL-6 MM cells via inhibition of an HGF autocrine growth loop and thymidine incorporation in INA-6 MM cells induced by exogenous HGF. HGF-induced migration and adhesion of INA-6 were completely and specifically blocked by the Nanobody. Furthermore, the Nanobody abolished the inhibiting effect of HGF on bone morphogenetic protein-2-induced alkaline phosphatase activity and the mineralization of human mesenchymal stem cells. Finally, we show that the Nanobody reduced phosphorylation of tyrosine residues in c-MET, MAPK, and Akt. We also compared the Nanobody with anti-c-MET monoclonal antibodies and revealed the similar or better effect. CONCLUSIONS The anti-c-MET Nanobody inhibited MM cell migration, thymidine incorporation, and adhesion, and blocked the HGF-mediated inhibition of osteoblastogenesis. The anti-c-MET Nanobody might represent a novel therapeutic agent in the treatment of MM and other cancers driven by HGF-c-MET signaling.
Collapse
Affiliation(s)
- Tobias Schmidt Slørdahl
- The KG Jebsen Center for Myeloma Research and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | | | | | | | | |
Collapse
|
105
|
Meyer AS, Miller MA, Gertler FB, Lauffenburger DA. The receptor AXL diversifies EGFR signaling and limits the response to EGFR-targeted inhibitors in triple-negative breast cancer cells. Sci Signal 2013; 6:ra66. [PMID: 23921085 DOI: 10.1126/scisignal.2004155] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The relationship between drug resistance, changes in signaling, and emergence of an invasive phenotype is well appreciated, but the underlying mechanisms are not well understood. Using machine learning analysis applied to the Cancer Cell Line Encyclopedia database, we identified expression of AXL, the gene that encodes the epithelial-to-mesenchymal transition (EMT)-associated receptor tyrosine kinase (RTK) AXL, as exceptionally predictive of lack of response to ErbB family receptor-targeted inhibitors. Activation of EGFR (epidermal growth factor receptor) transactivated AXL, and this ligand-independent AXL activity diversified EGFR-induced signaling into additional downstream pathways beyond those triggered by EGFR alone. AXL-mediated signaling diversification was required for EGF (epidermal growth factor)-elicited motility responses in AXL-positive TNBC (triple-negative breast cancer) cells. Using cross-linking coimmunoprecipitation assays, we determined that AXL associated with EGFR, other ErbB receptor family members, MET (hepatocyte growth factor receptor), and PDGFR (platelet-derived growth factor receptor) but not IGF1R (insulin-like growth factor 1 receptor) or INSR (insulin receptor). From these AXL interaction data, we predicted AXL-mediated signaling synergy for additional RTKs and validated these predictions in cells. This alternative mechanism of receptor activation limits the use of ligand-blocking therapies and indicates against therapy withdrawal after acquired resistance. Further, subadditive interaction between EGFR- and AXL-targeted inhibitors across all AXL-positive TNBC cell lines may indicate that increased abundance of EGFR is principally a means to transactivation-mediated signaling.
Collapse
Affiliation(s)
- Aaron S Meyer
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | |
Collapse
|
106
|
Xiang H, Bender BC, Reyes AE, Merchant M, Jumbe NL', Romero M, Davancaze T, Nijem I, Mai E, Young J, Peterson A, Damico-Beyer LA. Onartuzumab (MetMAb): using nonclinical pharmacokinetic and concentration-effect data to support clinical development. Clin Cancer Res 2013; 19:5068-78. [PMID: 23894056 DOI: 10.1158/1078-0432.ccr-13-0260] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We characterized the pharmacokinetics of onartuzumab (MetMAb) in animals and determined a concentration-effect relationship in tumor-bearing mice to enable estimation of clinical pharmacokinetics and target doses. EXPERIMENTAL DESIGN A tumor growth inhibition model was used to estimate tumoristatic concentrations (TSC) in mice. Human pharmacokinetic parameters were projected from pharmacokinetics in cynomolgus monkeys by the species-invariant time method. Monte Carlo simulations predicted the percentage of patients achieving steady-state trough serum concentrations (Ctrough ss) ≥TSC for every 3-week (Q3W) dosing. RESULTS Onartuzumab clearance (CL) in the linear dose range was 21.1 and 12.2 mL/d/kg in mice and cynomolgus monkeys with elimination half-life at 6.10 and 3.37 days, respectively. The estimated TSC in KP4 pancreatic xenograft tumor-bearing mice was 15 μg/mL. Projected CL for humans in the linear dose range was 5.74 to 9.36 mL/d/kg with scaling exponents of CL at 0.75 to 0.9. Monte Carlo simulations projected a Q3W dose of 10 to 30 mg/kg to achieve Ctrough ss of 15 μg/mL in 95% or more of patients. CONCLUSIONS Onartuzumab pharmacokinetics differed from typical bivalent glycosylated monoclonal antibodies with approximately 2-times faster CL in the linear dose range. Despite this higher CL, xenograft efficacy data supported dose flexibility with Q1W to Q3W dose regimens in the clinical setting with a TSC of 15 μg/mL as the Ctrough ss target. The projected human efficacious dose of 10 to 30 mg/kg Q3W should achieve the target TSC of 15 μg/mL. These data show effective pharmacokinetic/pharmacodynamic modeling to project doses to be tested in the clinic.
Collapse
Affiliation(s)
- Hong Xiang
- Authors' Affiliations: Departments of Pharmacokinetic and Pharmacodynamic Sciences, Translational Oncology, Bioanalytical Sciences, Biochemical and Cellular Pharmacology, and Portfolio Management and Operations, Genentech, Inc., South San Francisco; Quantitative Solutions, Menlo Park; Medivation, Inc., San Francisco; and Celgene, San Diego, California
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Monovalent antibody design and mechanism of action of onartuzumab, a MET antagonist with anti-tumor activity as a therapeutic agent. Proc Natl Acad Sci U S A 2013; 110:E2987-96. [PMID: 23882082 DOI: 10.1073/pnas.1302725110] [Citation(s) in RCA: 199] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Binding of hepatocyte growth factor (HGF) to the receptor tyrosine kinase MET is implicated in the malignant process of multiple cancers, making disruption of this interaction a promising therapeutic strategy. However, targeting MET with bivalent antibodies can mimic HGF agonism via receptor dimerization. To address this limitation, we have developed onartuzumab, an Escherichia coli-derived, humanized, and affinity-matured monovalent monoclonal antibody against MET, generated using the knob-into-hole technology that enables the antibody to engage the receptor in a one-to-one fashion. Onartuzumab potently inhibits HGF binding and receptor phosphorylation and signaling and has antibody-like pharmacokinetics and antitumor activity. Biochemical data and a crystal structure of a ternary complex of onartuzumab antigen-binding fragment bound to a MET extracellular domain fragment, consisting of the MET Sema domain fused to the adjacent Plexins, Semaphorins, Integrins domain (MET Sema-PSI), and the HGF β-chain demonstrate that onartuzumab acts specifically by blocking HGF α-chain (but not β-chain) binding to MET. These data suggest a likely binding site of the HGF α-chain on MET, which when dimerized leads to MET signaling. Onartuzumab, therefore, represents the founding member of a class of therapeutic monovalent antibodies that overcomes limitations of antibody bivalency for targets impacted by antibody crosslinking.
Collapse
|
108
|
Abstract
PURPOSE OF REVIEW The mesenchymal-epidermal transition (c-MET) receptor tyrosine kinase has a central role in the cancer cell's survival. MET and its ligand, hepatocyte growth factor (HGF), have recently been identified as promising targets in solid tumors, including nonsmall-cell lung cancer (NSCLC). RECENT FINDINGS Aberrant MET activation can be the result of different mechanisms such as MET and HGF overexpression, MET gene amplification or mutation. Retrospective studies in NSCLC showed that MET gene copy number is a negative prognostic factor, although few data are available on the role of MET mutations. In preclinical models, cell lines with MET gene amplification are extremely sensitive to MET inhibition. Although the inner presence of gene amplification is a rare event (1-7% cases), MET amplification has emerged as one of the critical events for acquired resistance in epidermal growth factor receptor (EGFR) mutated lung adenocarcinomas refractory to EGFR-tyrosine kinase inhibitors (TKIs). In NSCLC with acquired resistance to EGFR-TKIs, MET amplification occurs in up to 20% cases and preclinical and clinical data indicated MET and EGFR co-inhibition as a potential effective strategy to overcome resistance. SUMMARY MET has recently emerged as a promising target, and ongoing trials will clarify the role of anti-MET strategies in NSCLC.
Collapse
|
109
|
Von Kreudenstein TS, Escobar-Carbrera E, Lario PI, D'Angelo I, Brault K, Kelly J, Durocher Y, Baardsnes J, Woods RJ, Xie MH, Girod PA, Suits MDL, Boulanger MJ, Poon DKY, Ng GYK, Dixit SB. Improving biophysical properties of a bispecific antibody scaffold to aid developability: quality by molecular design. MAbs 2013; 5:646-54. [PMID: 23924797 DOI: 10.4161/mabs.25632] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
While the concept of Quality-by-Design is addressed at the upstream and downstream process development stages, we questioned whether there are advantages to addressing the issues of biologics quality early in the design of the molecule based on fundamental biophysical characterization, and thereby reduce complexities in the product development stages. Although limited number of bispecific therapeutics are in clinic, these developments have been plagued with difficulty in producing materials of sufficient quality and quantity for both preclinical and clinical studies. The engineered heterodimeric Fc is an industry-wide favorite scaffold for the design of bispecific protein therapeutics because of its structural, and potentially pharmacokinetic, similarity to the natural antibody. Development of molecules based on this concept, however, is challenged by the presence of potential homodimer contamination and stability loss relative to the natural Fc. We engineered a heterodimeric Fc with high heterodimeric specificity that also retains natural Fc-like biophysical properties, and demonstrate here that use of engineered Fc domains that mirror the natural system translates into an efficient and robust upstream stable cell line selection process as a first step toward a more developable therapeutic.
Collapse
|
110
|
Spiess C, Merchant M, Huang A, Zheng Z, Yang NY, Peng J, Ellerman D, Shatz W, Reilly D, Yansura DG, Scheer JM. Bispecific antibodies with natural architecture produced by co-culture of bacteria expressing two distinct half-antibodies. Nat Biotechnol 2013; 31:753-8. [DOI: 10.1038/nbt.2621] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 05/20/2013] [Indexed: 11/09/2022]
|
111
|
Landi L, Minuti G, D’Incecco A, Salvini J, Cappuzzo F. MET overexpression and gene amplification in NSCLC: a clinical perspective. LUNG CANCER (AUCKLAND, N.Z.) 2013; 4:15-25. [PMID: 28210131 PMCID: PMC5217438 DOI: 10.2147/lctt.s35168] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transmembrane tyrosine kinase mesenchymal-epidermal transition (MET) receptor and its ligand, hepatocyte growth factor, also known as scatter factor, have recently been identified as novel promising targets in several human malignancies, including non-small cell lung cancer (NSCLC). Amplification, mutation, or overexpression of the MET gene can result in aberrant activation of the MET axis, leading to migration, invasion, proliferation, metastasis, and neoangiogenesis of cancer cells, suggesting that interfering with the MET/hepatocyte growth factor pathway could represent a potential antitumor strategy. While the role of MET mutations in NSCLC is not as yet fully understood, retrospective studies have shown that an increased MET gene copy number is a negative prognostic factor. In NSCLC, amplification of the MET gene is a relatively rare event, occurring in approximately 4% of patients not previously exposed to systemic therapies and in up to 20% of patients with acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors. In preclinical models, the presence of MET amplification is a predictor of high sensitivity to anti-MET compounds, and several agents have entered in clinical trials for patients having advanced disease, with promising results. The aim of the present review is to summarize available data on the role of MET in NSCLC and to describe therapeutic strategies under investigation.
Collapse
Affiliation(s)
- Lorenza Landi
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| | - Gabriele Minuti
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| | - Armida D’Incecco
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| | - Jessica Salvini
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| | - Federico Cappuzzo
- Medical Oncology Department, Istituto Toscano Tumori, Ospedale Civile, Livorno, Italy
| |
Collapse
|
112
|
Giroux Leprieur É. [A new drug in thoracic oncology: MetMab (onartuzumab)]. REVUE DE PNEUMOLOGIE CLINIQUE 2013; 69:152-158. [PMID: 23477747 DOI: 10.1016/j.pneumo.2012.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 10/29/2012] [Accepted: 11/06/2012] [Indexed: 06/01/2023]
Abstract
Met pathway is activated in many solid cancers. In advanced non-small cell lung cancer (NSCLC), Met amplification is involved in 5 to 20% of acquired resistance to EGFR tyrosine kinase inhibitors (TKI) in tumors with initially sensitive EGFR mutation. MetMab (onartuzumab) is a monoclonal single-arm humanized anti-Met antibody. Its fixation on the Met receptor prevents the binding of the ligand (Hepatocyte Growth factor [HGF]) and the signal transduction. After promising results in preclinical and phase I trials, a randomized phase II trial has been conducted in advanced NSCLC in 2nd or 3rd line treatment. One hundred and twenty-eight patients have been randomized between an association of erlotinib+placebo and erlotinib+MetMab (15mg/kg IV every 3 weeks) until progression or toxicity. Patients with overexpression of Met in immunohistochemistry (IHC) had a progression-free survival (PFS) and an overall survival (OS) two-fold (median 1.5 versus 2.9 months; HR=0.53; P=0.04) and three-fold (median 3.8 versus 12.6 months; HR=0.37; P=0.002) longer, respectively, than patients with negative IHC score. The erlotinib+MetMab association had a worse effect on SSP and OS than the control arm in patients with negative IHC. The toxicity profile of MetMab is very good, and the main adverse effect is the occurrence of peripheral edemas, most of the time of low grade. A randomized phase III is on going to validate these results.
Collapse
Affiliation(s)
- É Giroux Leprieur
- Service de pneumologie et oncologie thoracique, université Versailles-Saint-Quentin-en-Yvelines, hôpital Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92100 Boulogne-Billancourt, France.
| |
Collapse
|
113
|
|
114
|
Diamond JR, Salgia R, Varella-Garcia M, Kanteti R, LoRusso PM, Clark JW, Xu LG, Wilner K, Eckhardt SG, Ching KA, Lira ME, Schoenmakers EFPM, Christensen JG, Camidge DR. Initial clinical sensitivity and acquired resistance to MET inhibition in MET-mutated papillary renal cell carcinoma. J Clin Oncol 2013; 31:e254-8. [PMID: 23610116 DOI: 10.1200/jco.2012.46.4289] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
115
|
Tsao AS, Papadimitrakopoulou V. The importance of molecular profiling in predicting response to epidermal growth factor receptor family inhibitors in non-small-cell lung cancer: focus on clinical trial results. Clin Lung Cancer 2013; 14:311-21. [PMID: 23582282 DOI: 10.1016/j.cllc.2013.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 01/16/2013] [Accepted: 01/29/2013] [Indexed: 01/28/2023]
Abstract
In recent years, the epidermal growth factor receptor (EGFR) family has become a key focus of non-small-cell lung cancer biology and targeted therapies, such as the reversible EGFR tyrosine kinase inhibitors erlotinib and gefitinib. Initially, response to these agents was associated with certain demographic and clinical characteristics; subsequently, it was discovered that these subgroups were more likely to harbor specific mutations in the EGFR gene that enhanced tumor response. However, the presence of these mutations does not equate to therapeutic success. Other aspects of EGFR family signaling, including other types of EGFR mutations, EGFR protein expression, EGFR gene amplification, mediators of downstream signaling, and other receptors with similar downstream pathways may all play a role in response or resistance to treatment. The identification of these and other molecular determinants is driving the development of novel therapies designed to achieve improved clinical outcomes in patients.
Collapse
Affiliation(s)
- Anne S Tsao
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | |
Collapse
|
116
|
Sennino B, Ishiguro-Oonuma T, Schriver BJ, Christensen JG, McDonald DM. Inhibition of c-Met reduces lymphatic metastasis in RIP-Tag2 transgenic mice. Cancer Res 2013; 73:3692-703. [PMID: 23576559 DOI: 10.1158/0008-5472.can-12-2160] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inhibition of VEGF signaling can promote lymph node metastasis in preclinical models, but the mechanism is not fully understood, and successful methods of prevention have not been found. Signaling of hepatocyte growth factor (HGF) and its receptor c-Met can promote the growth of lymphatics and metastasis of some tumors. We sought to explore the contributions of c-Met signaling to lymph node metastasis after inhibition of VEGF signaling. In particular, we examined whether c-Met is upregulated in lymphatics in or near pancreatic neuroendocrine tumors in RIP-Tag2 transgenic mice and whether lymph node metastasis can be reduced by concurrent inhibition of VEGF and c-Met signaling. Inhibition of VEGF signaling by anti-VEGF antibody or sunitinib in mice from the age of 14 to 17 weeks was accompanied by more intratumoral lymphatics, more tumor cells inside lymphatics, and more lymph node metastases. Under these conditions, lymphatic endothelial cells, like tumor cells, had strong immunoreactivity for c-Met and phospho-c-Met. c-Met blockade by the selective inhibitor, PF-04217903, significantly reduced metastasis to local lymph nodes. Together, these results indicate that inhibition of VEGF signaling in RIP-Tag2 mice upregulates c-Met expression in lymphatic endothelial cells, increases the number of intratumoral lymphatics and number of tumor cells within lymphatics, and promotes metastasis to local lymph nodes. Prevention of lymph node metastasis by PF-04217903 in this setting implicates c-Met signaling in tumor cell spread to lymph nodes.
Collapse
Affiliation(s)
- Barbara Sennino
- Department of Anatomy, Comprehensive Cancer Center, Cardiovascular Research Institute, University of California-San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
117
|
Bladt F, Faden B, Friese-Hamim M, Knuehl C, Wilm C, Fittschen C, Grädler U, Meyring M, Dorsch D, Jaehrling F, Pehl U, Stieber F, Schadt O, Blaukat A. EMD 1214063 and EMD 1204831 constitute a new class of potent and highly selective c-Met inhibitors. Clin Cancer Res 2013; 19:2941-51. [PMID: 23553846 DOI: 10.1158/1078-0432.ccr-12-3247] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The mesenchymal-epithelial transition factor (c-Met) receptor, also known as hepatocyte growth factor receptor (HGFR), controls morphogenesis, a process that is physiologically required for embryonic development and tissue repair. Aberrant c-Met activation is associated with a variety of human malignancies including cancers of the lung, kidney, stomach, liver, and brain. In this study, we investigated the properties of two novel compounds developed to selectively inhibit the c-Met receptor in antitumor therapeutic interventions. EXPERIMENTAL DESIGN The pharmacologic properties, c-Met inhibitory activity, and antitumor effects of EMD 1214063 and EMD 1204831 were investigated in vitro and in vivo, using human cancer cell lines and mouse xenograft models. RESULTS EMD 1214063 and EMD 1204831 selectively suppressed the c-Met receptor tyrosine kinase activity. Their inhibitory activity was potent [inhibitory 50% concentration (IC50), 3 nmol/L and 9 nmol/L, respectively] and highly selective, when compared with their effect on a panel of 242 human kinases. Both EMD 1214063 and EMD 1204831 inhibited c-Met phosphorylation and downstream signaling in a dose-dependent fashion, but differed in the duration of their inhibitory activity. In murine xenograft models, both compounds induced regression of human tumors, regardless of whether c-Met activation was HGF dependent or independent. Both drugs were well tolerated and induced no substantial weight loss after more than 3 weeks of treatment. CONCLUSIONS Our results indicate selective c-Met inhibition by EMD 1214063 and EMD 1204831 and strongly support clinical testing of these compounds in the context of molecularly targeted anticancer strategies.
Collapse
Affiliation(s)
- Friedhelm Bladt
- EMD Serono and Merck Serono Research and Development, Merck KGaA, Darmstadt, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Penuel E, Li C, Parab V, Burton L, Cowan KJ, Merchant M, Yauch RL, Patel P, Peterson A, Hampton GM, Lackner MR, Hegde PS. HGF as a circulating biomarker of onartuzumab treatment in patients with advanced solid tumors. Mol Cancer Ther 2013; 12:1122-30. [PMID: 23536720 DOI: 10.1158/1535-7163.mct-13-0015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The objective of this study was to evaluate circulating hepatocyte growth factor (cHGF) as a pharmacodynamic biomarker of Met inhibition for onartuzumab (MetMAb, OA5D5v2) in a phase I trial in patients with advanced cancers and a phase II trial in non-small cell lung cancer (NSCLC). The phase I study was a dose escalation trial with onartuzumab administered i.v. once every three weeks. The phase II study was a randomized two-arm trial in which onartuzumab or placebo was administered in combination with erlotinib in 137 patients with second and third line (2/3L) NSCLC. cHGF levels were evaluated by ELISA at multiple time points over the treatment period. Onartuzumab administration resulted in an acute and sustained rise in cHGF in both the phase I and phase II studies. Elevation in cHGF was independent of dose or drug exposure and was restricted to onartuzumab treatment. Neither higher baseline nor elevated change in cHGF levels upon treatment could simply be attributed to tumor burden or number of liver metastasis. We have shown that elevated cHGF can consistently and reproducibly be measured as a pharmacodynamic biomarker of onartuzumab activity. The elevation in cHGF is independent of tumor type, dose administered, or dose duration. Although these studies were not powered to directly address the contribution of cHGF as a predictive, on-treatment, circulating biomarker, these data suggest that measurement of cHGF in future expanded studies is warranted.
Collapse
|
119
|
Abstract
Lung cancer is a heterogeneous group of disorders that is now being subdivided into molecular subtypes with dedicated targeted therapies. The MET receptor tyrosine kinase has been identified as aberrantly overexpressed, potentially having activating mutations, and amplified in certain subsets of lung cancers. The ligand hepatocyte growth factor (HGF) can also be overexpressed in lung cancer or expressed in stroma, and both the MET receptor and the HGF ligand can be targets for therapeutics, especially in lung cancer. Activation of MET leads to a plethora of biochemical and biologic changes both in normal and cancerous cells. Preclinically, it has been shown that silencing or inactivating MET leads to decreased viability of cancer cells. There are a number of compounds against MET/HGF in clinical trials that have been shown to be active in lung cancers. This review will summarize the biology of MET as well as its therapeutic inhibition in lung cancer.
Collapse
Affiliation(s)
| | - Ravi Salgia
- All authors: University of Chicago, Chicago, IL
| |
Collapse
|
120
|
Zheng X, He K, Zhang L, Yu J. Crizotinib induces PUMA-dependent apoptosis in colon cancer cells. Mol Cancer Ther 2013; 12:777-86. [PMID: 23427294 DOI: 10.1158/1535-7163.mct-12-1146] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Oncogenic alterations in MET or anaplastic lymphoma kinase (ALK) have been identified in a variety of human cancers. Crizotinib (PF02341066) is a dual MET and ALK inhibitor and approved for the treatment of a subset of non-small cell lung carcinoma and in clinical development for other malignancies. Crizotinib can induce apoptosis in cancer cells, whereas the underlying mechanisms are not well understood. In this study, we found that crizotinib induces apoptosis in colon cancer cells through the BH3-only protein PUMA. In cells with wild-type p53, crizotinib induces rapid induction of PUMA and Bim accompanied by p53 stabilization and DNA damage response. The induction of PUMA and Bim is mediated largely by p53, and deficiency in PUMA or p53, but not Bim, blocks crizotinib-induced apoptosis. Interestingly, MET knockdown led to selective induction of PUMA, but not Bim or p53. Crizotinib also induced PUMA-dependent apoptosis in p53-deficient colon cancer cells and synergized with gefitinib or sorafenib to induce marked apoptosis via PUMA in colon cancer cells. Furthermore, PUMA deficiency suppressed apoptosis and therapeutic responses to crizotinib in xenograft models. These results establish a critical role of PUMA in mediating apoptotic responses of colon cancer cells to crizotinib and suggest that mechanisms of oncogenic addiction to MET/ALK-mediated survival may be cell type-specific. These findings have important implications for future clinical development of crizotinib.
Collapse
Affiliation(s)
- Xingnan Zheng
- Department of Pathology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | | | | | | |
Collapse
|
121
|
Broggini M, Garassino MC, Damia G. Evaluation of safety and efficacy of tivantinib in the treatment of inoperable or recurrent non-small-cell lung cancer. Cancer Manag Res 2013; 5:15-20. [PMID: 23378782 PMCID: PMC3559079 DOI: 10.2147/cmar.s29995] [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] [Indexed: 11/28/2022] Open
Abstract
Tivantinib is a selective, oral, non-ATP-competitive, small molecule inhibitor of the c-Met receptor, tyrosine kinase, which is implicated at different levels of tumor cell migration, invasion, proliferation, and metastasis. Tivantinib has shown antitumor activity in various human tumor cell lines and in xenograft models of human cancers, including non-small-cell lung cancer. Few therapeutic options are available at present for advanced non-small-cell lung cancer, so there is a pressing need for new therapeutic strategies to improve response and survival. Amplification of Met has been reported in more than 20% of lung tumors that have acquired resistance to epidermal growth factor receptor inhibitors, implying that treatment of these tumors with a c-Met inhibitor should overcome resistance. Tivantinib has shown interesting and promising results in advanced non-small-cell lung cancer and appears to be well tolerated, either alone or in combination with other drugs. An interesting additional feature is the ability of the drug to delay development of new metastasis, in agreement with the proposed role of Met in this particular setting.
Collapse
Affiliation(s)
- Massimo Broggini
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | | | | |
Collapse
|
122
|
Lee JM, Kim B, Lee SB, Jeong Y, Oh YM, Song YJ, Jung S, Choi J, Lee S, Cheong KH, Kim DU, Park HW, Han YK, Kim GW, Choi H, Song PH, Kim KA. Cbl-independent degradation of Met: ways to avoid agonism of bivalent Met-targeting antibody. Oncogene 2012. [DOI: 10.1038/onc.2012.551] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
123
|
Oh YM, Song YJ, Lee SB, Jeong Y, Kim B, Kim GW, Kim KE, Lee JM, Cho MY, Choi J, Nam DH, Song PH, Cheong KH, Kim KA. A new anti-c-Met antibody selected by a mechanism-based dual-screening method: therapeutic potential in cancer. Mol Cells 2012; 34:523-9. [PMID: 23180291 PMCID: PMC3887825 DOI: 10.1007/s10059-012-0194-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 09/11/2012] [Accepted: 10/17/2012] [Indexed: 10/27/2022] Open
Abstract
c-Met, the high affinity receptor for hepatocyte growth factor (HGF), is one of the most frequently activated tyrosine kinases in many human cancers and a target for cancer therapy. However, inhibitory targeting of c-Met with antibodies has proven difficult, because most antibodies have intrinsic agonist activity. Therefore, the strategy for reducing the agonism is critical for successful development of cancer therapies based on anti-c-Met antibodies. Here we developed a mechanism-based assay method for rapid screening of anti-c-Met antibodies, involving the determination of Akt phosphorylation and c-Met degradation for agonism and efficacy, respectively. Using the method, we identified an antibody, F46, that binds to human c-Met with high affinity (Kd = 2.56 nM) and specificity, and induces the degradation of c-Met in multiple cancer cells (including MKN45, a gastric cancer cell line) with minimal activation of c-Met signaling. F46 induced c-Met internalization in both HGF-dependent and HGF-independent cells, suggesting that the degradation of c-Met results from antibody-mediated receptor internalization. Furthermore, F46 competed with HGF for binding to c-Met, resulting in the inhibition of both HGF-mediated invasion and angiogenesis. Consistently, F46 inhibited the proliferation of MKN45 cells, in which c-Met is constitutively activated in an HGF-independent manner. Xenograft analysis revealed that F46 markedly inhibits the growth of subcutaneously implanted gastric and lung tumors. These results indicate that F46, identified by a novel mechanism-based assay, induces c-Met degradation with minimal agonism, implicating a potential role of F46 in therapy of human cancers.
Collapse
Affiliation(s)
- Young Mi Oh
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Yun-Jeong Song
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Saet Byoul Lee
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Yunju Jeong
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Bogyou Kim
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Geun Woong Kim
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Kyung Eun Kim
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Ji Min Lee
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Mi-Young Cho
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Jaehyun Choi
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | | | - Paul H Song
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Kwang Ho Cheong
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| | - Kyung-Ah Kim
- Therapeutic Antibody Group, Samsung Advanced Institute of Technology, Yongin 446-712,
Korea
| |
Collapse
|
124
|
Feng Y, Ma PC. Anti-MET targeted therapy has come of age: the first durable complete response with MetMAb in metastatic gastric cancer. Cancer Discov 2012; 1:550-4. [PMID: 22586678 DOI: 10.1158/2159-8290.cd-11-0289] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The MET/hepatocyte growth factor (HGF) signaling pathway plays important roles in oncogenesis and tumor progression in a variety of human cancers. MET/HGF drives an invasive signaling program that can be dysregulated in human cancers through a number of activating mechanisms, including mutations, overexpression, amplification, alternative splicing, and HGF ligand-induced autocrine/paracrine loop signaling. As a testimony of MET-targeting therapeutics is beginning to come to clinical fruition, Catenacci and colleagues report the first case of durable complete response under an anti-MET receptor monoclonal antibody, MetMAb, in a patient with chemotherapy-refractory, advanced gastric cancer metastatic to the liver, found to have high MET gene polysomy and remarkably high serum HGF level. Serum and tissue studies also revealed predictive biomarkers for therapeutic response to MET inhibition.
Collapse
Affiliation(s)
- Yan Feng
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio 44195, USA
| | | |
Collapse
|
125
|
Fasolo A, Sessa C, Gianni L, Broggini M. Seminars in clinical pharmacology: an introduction to MET inhibitors for the medical oncologist. Ann Oncol 2012; 24:14-20. [PMID: 23110808 DOI: 10.1093/annonc/mds520] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
MET is a tyrosine kinase receptor for hepatocyte growth factor (HGF), primarily expressed on epithelial cells; the activation of MET induces several biological responses relevant for the development and growth of many human cancers. Several human malignancies present altered expression of MET and this is usually associated with poor prognosis and aggressive phenotype. The majority of MET inhibitors in clinical development target directly the receptor through the use of monoclonal antibodies (MAbs) or through small molecule inhibitors of MET kinase activity; small molecule inhibitors are very potent but less specific than MAbs. MET inhibitors are of great clinical interest because of the extensive crosstalk of the HGF/MET axis with many other signaling pathways, including growth factor-dependent pathways (like PI3K/AKT/mTOR,RAS/RAF/ERK) and vascular endothelial growth factor (VEGF) axis. In preclinical studies, the treatment with MET inhibitors could prevent or reverse resistance to inhibitors of growth factor-dependent signaling; this hypothesis is currently tested in phase III trials with anti-epidermal growth factor receptor (EGFR) inhibitors in non-small-cell lung cancer (NSCLC). Based on preclinical and preliminary clinical results, a rational strategy for the clinical development of MET antagonists should include a selection of the tumors with MET overexpression, the identification of prognostic/predictive biomarkers, the evaluation of combinations with anti-VEGF compounds.
Collapse
Affiliation(s)
- A Fasolo
- San Raffaele Hospital, IRCCS, Unit of New Drugs & Innovative Therapies, Department of Medical Oncology, Milan, Italy
| | | | | | | |
Collapse
|
126
|
Michaud NR, Jani JP, Hillerman S, Tsaparikos KE, Barbacci-Tobin EG, Knauth E, Putz H, Campbell M, Karam GA, Chrunyk B, Gebhard DF, Green LL, Xu JJ, Dunn MC, Coskran TM, Lapointe JM, Cohen BD, Coleman KG, Bedian V, Vincent P, Kajiji S, Steyn SJ, Borzillo GV, Los G. Biochemical and pharmacological characterization of human c-Met neutralizing monoclonal antibody CE-355621. MAbs 2012; 4:710-23. [PMID: 23007574 DOI: 10.4161/mabs.22160] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The c-Met proto-oncogene is a multifunctional receptor tyrosine kinase that is stimulated by its ligand, hepatocyte growth factor (HGF), to induce cell growth, motility and morphogenesis. Dysregulation of c-Met function, through mutational activation or overexpression, has been observed in many types of cancer and is thought to contribute to tumor growth and metastasis by affecting mitogenesis, invasion, and angiogenesis. We identified human monoclonal antibodies that bind to the extracellular domain of c-Met and inhibit tumor growth by interfering with ligand-dependent c-Met activation. We identified antibodies representing four independent epitope classes that inhibited both ligand binding and ligand-dependent activation of c-Met in A549 cells. In cells, the antibodies antagonized c-Met function by blocking receptor activation and by subsequently inducing downregulation of the receptor, translating to phenotypic effects in soft agar growth and tubular morphogenesis assays. Further characterization of the antibodies in vivo revealed significant inhibition of c-Met activity (≥ 80% lasting for 72-96 h) in excised tumors corresponded to tumor growth inhibition in multiple xenograft tumor models. Several of the antibodies identified inhibited the growth of tumors engineered to overexpress human HGF and human c-Met (S114 NIH 3T3) when grown subcutaneously in athymic mice. Furthermore, lead candidate antibody CE-355621 inhibited the growth of U87MG human glioblastoma and GTL-16 gastric xenografts by up to 98%. The findings support published pre-clinical and clinical data indicating that targeting c-Met with human monoclonal antibodies is a promising therapeutic approach for the treatment of cancer.
Collapse
Affiliation(s)
- Neil R Michaud
- Pfizer Global Research and Development, Groton, CT, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Luraghi P, Schelter F, Krüger A, Boccaccio C. The MET Oncogene as a Therapeutical Target in Cancer Invasive Growth. Front Pharmacol 2012; 3:164. [PMID: 22973229 PMCID: PMC3438853 DOI: 10.3389/fphar.2012.00164] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/21/2012] [Indexed: 01/30/2023] Open
Abstract
The MET proto-oncogene, encoding the tyrosine kinase receptor for Hepatocyte Growth Factor (HGF) regulates invasive growth, a genetic program that associates control of cell proliferation with invasion of the extracellular matrix and protection from apoptosis. Physiologically, invasive growth takes place during embryonic development, and, in post-natal life, in wound healing and regeneration of several tissues. The MET oncogene is overexpressed and/or genetically mutated in many tumors, thereby sustaining pathological invasive growth, a prerequisite for metastasis. MET is the subject of intense research as a target for small molecule kinase inhibitors and, together with its ligand HGF, for inhibitory antibodies. The tight interplay of MET with the protease network has unveiled mechanisms to be exploited to achieve effective inhibition of invasive growth.
Collapse
Affiliation(s)
- Paolo Luraghi
- Division of Experimental Clinical Molecular Oncology, IRCC – Institute for Cancer Research and Treatment, University of Turin Medical SchoolCandiolo, Italy
| | - Florian Schelter
- Klinikum rechts der Isar der Technischen Universität München, Institut für Experimentelle Onkologie und TherapieforschungMünchen, Germany
| | - Achim Krüger
- Klinikum rechts der Isar der Technischen Universität München, Institut für Experimentelle Onkologie und TherapieforschungMünchen, Germany
| | - Carla Boccaccio
- Division of Experimental Clinical Molecular Oncology, IRCC – Institute for Cancer Research and Treatment, University of Turin Medical SchoolCandiolo, Italy
| |
Collapse
|
128
|
Cui JJ, McTigue M, Nambu M, Tran-Dubé M, Pairish M, Shen H, Jia L, Cheng H, Hoffman J, Le P, Jalaie M, Goetz GH, Ryan K, Grodsky N, Deng YL, Parker M, Timofeevski S, Murray BW, Yamazaki S, Aguirre S, Li Q, Zou H, Christensen J. Discovery of a Novel Class of Exquisitely Selective Mesenchymal-Epithelial Transition Factor (c-MET) Protein Kinase Inhibitors and Identification of the Clinical Candidate 2-(4-(1-(Quinolin-6-ylmethyl)-1H-[1,2,3]triazolo[4,5-b]pyrazin-6-yl)-1H-pyrazol-1-yl)ethanol (PF-04217903) for the Treatment of Cancer. J Med Chem 2012; 55:8091-109. [DOI: 10.1021/jm300967g] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- J. Jean Cui
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Michele McTigue
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Mitchell Nambu
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Michelle Tran-Dubé
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Mason Pairish
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Hong Shen
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Lei Jia
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Hengmiao Cheng
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Jacqui Hoffman
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Phuong Le
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Mehran Jalaie
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Gilles H. Goetz
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Kevin Ryan
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Neil Grodsky
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Ya-li Deng
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Max Parker
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Sergei Timofeevski
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Brion W. Murray
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Shinji Yamazaki
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Shirley Aguirre
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Qiuhua Li
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Helen Zou
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - James Christensen
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| |
Collapse
|
129
|
Discovery of novel 2-aminopyridine-3-carboxamides as c-Met kinase inhibitors. Bioorg Med Chem 2012; 20:5169-80. [DOI: 10.1016/j.bmc.2012.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Revised: 07/03/2012] [Accepted: 07/04/2012] [Indexed: 12/21/2022]
|
130
|
Klein C, Sustmann C, Thomas M, Stubenrauch K, Croasdale R, Schanzer J, Brinkmann U, Kettenberger H, Regula JT, Schaefer W. Progress in overcoming the chain association issue in bispecific heterodimeric IgG antibodies. MAbs 2012; 4:653-63. [PMID: 22925968 DOI: 10.4161/mabs.21379] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The development of bispecific antibodies has attracted substantial interest, and many different formats have been described. Those specifically containing an Fc part are mostly tetravalent, such as stabilized IgG-scFv fusions or dual-variable domain (DVD) IgGs. However, although they exhibit IgG-like properties and technical developability, these formats differ in size and geometry from classical IgG antibodies. Thus, considerable efforts focus on bispecific heterodimeric IgG antibodies that more closely mimic natural IgG molecules. The inherent chain association problem encountered when producing bispecific heterodimeric IgG antibodies can be overcome by several methods. While technologies like knobs-into-holes (KiH) combined with a common light chain or the CrossMab technology enforce the correct chain association, other approaches, e.g., the dual-acting Fab (DAF) IgGs, do not rely on a heterodimeric Fc part. This review discusses the state of the art in bispecific heterodimeric IgG antibodies, with an emphasis on recent progress.
Collapse
Affiliation(s)
- Christian Klein
- Discovery Oncology, Roche Pharma Research and Early Development pRED, Roche Glycart AG, Schlieren, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Jagoda EM, Lang L, Bhadrasetty V, Histed S, Williams M, Kramer-Marek G, Mena E, Rosenblum L, Marik J, Tinianow JN, Merchant M, Szajek L, Paik C, Cecchi F, Raffensperger K, Jose-Dizon JM, Bottaro DP, Choyke P. Immuno-PET of the hepatocyte growth factor receptor Met using the 1-armed antibody onartuzumab. J Nucl Med 2012; 53:1592-600. [PMID: 22917884 DOI: 10.2967/jnumed.111.102293] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The overexpression and overactivation of hepatocyte growth factor receptor (Met) in various cancers has been linked to increased proliferation, progression to metastatic disease, and drug resistance. Developing a PET agent to assess Met expression would aid in the diagnosis and monitoring of responses to Met-targeted therapies. In these studies, onartuzumab, the experimental therapeutic 1-armed monoclonal antibody, was radiolabeled with (76)Br or (89)Zr and evaluated as an imaging agent in Met-expressing cell lines and mouse xenografts. METHODS (89)Zr-desferrioxamine (df)-onartuzumab was synthesized using a df-conjugate; (76)Br-onartuzumab was labeled directly. Met-binding studies were performed using the human tumor-derived cell lines MKN-45, SNU-16, and U87-MG, which have relatively high, moderate, and low levels of Met, respectively. Biodistribution and small-animal PET studies were performed in MKN-45 and U87-MG xenografts. RESULTS (76)Br-onartuzumab and (89)Zr-df-onartuzumab exhibited specific, high-affinity Met binding (in the nanomolar range) that was concordant with established Met expression levels. In MKN-45 (gastric carcinoma) xenografts, both tracers cleared slowly from nontarget tissues, with the highest uptake in tumor, blood, kidneys, and lungs. (76)Br-onartuzumab MKN-45 tumor uptake remained relatively constant from 18 h (5 percentage injected dose per gram of tissue [%ID/g]) to 48 h (3 %ID/g) and exhibited tumor-to-muscle ratios ranging from 4:1 to 6:1. In contrast, (89)Zr-df-onartuzumab MKN-45 tumor uptake continued to accumulate from 18 h (10 %ID/g) to 120 h (23 %ID/g), attaining tumor-to-muscle ratios ranging from 20:1 to 27:1. MKN-45 tumors were easily visualized in imaging studies with both tracers at 18 h, but after 48 h (89)Zr-df-onartuzumab image quality improved, with at least 2-fold-greater tumor uptake than nontarget tissues. MKN-45 tumor uptake for both tracers correlated significantly with tumor mass and Met expression and was not affected by the presence of plasma shed Met. CONCLUSION (89)Zr-df-onartuzumab and (76)Br-onartuzumab specifically targeted Met in vitro and in vivo; (89)Zr-df-onartuzumab achieved higher tumor uptake and tumor-to-muscle ratios than (76)Br-onartuzumab at later times, suggesting that (89)Zr-df-onartuzumab would be better suited to image Met for diagnostic and prognostic purposes.
Collapse
Affiliation(s)
- Elaine M Jagoda
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Abstract
The receptor tyrosine kinase c-MET and its ligand, hepatocyte growth factor (HGF), regulate multiple cellular processes that stimulate cell proliferation, invasion and angiogenesis. This review provides an overview of the evidence to support c-MET or the HGF/c-MET signaling pathway as relevant targets for personalized cancer treatment based on high frequencies of c-MET and/or HGF overexpression, activation, amplification in non-small cell lung carcinoma (NSCLC), gastric, ovarian, pancreatic, thyroid, breast, head and neck, colon and kidney carcinomas. Additionally, the current knowledge of small molecule inhibitors (tivantinib [ARQ 197]), c-MET/HGF antibodies (rilotumumab and MetMAb) and mechanisms of resistance to c-MET-targeted therapies are discussed.
Collapse
Affiliation(s)
- J Rafael Sierra
- Princess Margaret Hospital/Ontario Cancer Institute and University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
133
|
Sharma N, Adjei AA. In the clinic: ongoing clinical trials evaluating c-MET-inhibiting drugs. Ther Adv Med Oncol 2012; 3:S37-50. [PMID: 22128287 DOI: 10.1177/1758834011423403] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The c-MET (mesenchymal-epithelial transition factor) pathway is dysregulated in many human cancers and promotes tumor growth, invasion and dissemination. The c-MET receptor tyrosine kinase can be activated via gene mutation, gene amplification, protein overexpression and/or a ligand-dependent autocrine/paracrine loop. Abnormalities in c-MET signaling have been reported to correlate with poor clinical outcomes and drug resistance in patients with cancer. Significant progress has been made in advancement of c-MET pathway inhibitors through to clinical trials. A robust pipeline of high-quality inhibitors targeting different aspects of c-MET activation is currently being explored in phase I, II and III clinical trials across multiple tumor types. Preliminary data demonstrate promising clinical activity with these agents, along with an acceptable toxicity profile. In this manuscript, the pharmacological profile of drugs targeting the c-MET pathway and available data from ongoing clinical trials of these drugs are discussed.
Collapse
Affiliation(s)
- Neelesh Sharma
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
134
|
Belalcazar A, Azaña D, Perez CA, Raez LE, Santos ES. Targeting the Met pathway in lung cancer. Expert Rev Anticancer Ther 2012; 12:519-28. [PMID: 22500688 DOI: 10.1586/era.12.16] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Dysregulation of Met signaling has been implicated in the initiation, progression and metastasis of human cancers, and therefore represents an attractive target for anticancer drug development. Met is overexpressed in non-small-cell lung cancer and its lack of staining in normal lung tissue makes it an attractive target. To date, erlotinib and gefitinib have established themselves as first-line therapy for non-small-cell lung cancer patients whose tumors harbor an EGF receptor gene mutation, and hence, it is crucial that we identify mechanisms of resistance that could be targeted by novel agents, while keeping an acceptable toxicity profile at the same time; something very important when we develop these new drugs. Inhibitors of the Met pathway represent a therapeutic alternative in this setting. In this review, we discuss the early clinical studies reported using two Met inhibitors, a monoclonal antibody (MetMAb) and a small molecule tyrosine kinase inhibitor (MGCD265).
Collapse
Affiliation(s)
- Astrid Belalcazar
- Department of Internal Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | | | | |
Collapse
|
135
|
Bouzid K, Bedairia N, Marty M. Anticorps monoclonaux thérapeutiques en cancérologie. ACTA ACUST UNITED AC 2012; 60:223-8. [DOI: 10.1016/j.patbio.2012.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 04/25/2012] [Indexed: 10/28/2022]
|
136
|
Hill KS, Gaziova I, Harrigal L, Guerra YA, Qiu S, Sastry SK, Arumugam T, Logsdon CD, Elferink LA. Met receptor tyrosine kinase signaling induces secretion of the angiogenic chemokine interleukin-8/CXCL8 in pancreatic cancer. PLoS One 2012; 7:e40420. [PMID: 22815748 PMCID: PMC3398924 DOI: 10.1371/journal.pone.0040420] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 06/06/2012] [Indexed: 12/19/2022] Open
Abstract
At diagnosis, the majority of pancreatic cancer patients present with advanced disease when curative resection is no longer feasible and current therapeutic treatments are largely ineffective. An improved understanding of molecular targets for effective intervention of pancreatic cancer is thus urgent. The Met receptor tyrosine kinase is one candidate implicated in pancreatic cancer. Notably, Met is over expressed in up to 80% of invasive pancreatic cancers but not in normal ductal cells correlating with poor overall patient survival and increased recurrence rates following surgical resection. However the functional role of Met signaling in pancreatic cancer remains poorly understood. Here we used RNA interference to directly examine the pathobiological importance of increased Met signaling for pancreatic cancer. We show that Met knockdown in pancreatic tumor cells results in decreased cell survival, cell invasion, and migration on collagen I in vitro. Using an orthotopic model for pancreatic cancer, we provide in vivo evidence that Met knockdown reduced tumor burden correlating with decreased cell survival and tumor angiogenesis, with minimal effect on cell growth. Notably, we report that Met signaling regulates the secretion of the pro-angiogenic chemokine interleukin-8/CXCL8. Our data showing that the interleukin-8 receptors CXCR1 and CXCR2 are not expressed on pancreatic tumor cells, suggests a paracrine mechanism by which Met signaling regulates interleukin-8 secretion to remodel the tumor microenvironment, a novel finding that could have important clinical implications for improving the effectiveness of treatments for pancreatic cancer.
Collapse
Affiliation(s)
- Kristen S. Hill
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ivana Gaziova
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lindsay Harrigal
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yvette A. Guerra
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Suimin Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- UTMB Cancer Center, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Sarita K. Sastry
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- UTMB Cancer Center, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thiruvengadam Arumugam
- Department of Cancer Biology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Craig D. Logsdon
- Department of Cancer Biology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Lisa A. Elferink
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- UTMB Cancer Center, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
137
|
Yano S, Takeuchi S, Nakagawa T, Yamada T. Ligand-triggered resistance to molecular targeted drugs in lung cancer: roles of hepatocyte growth factor and epidermal growth factor receptor ligands. Cancer Sci 2012; 103:1189-94. [PMID: 22435662 DOI: 10.1111/j.1349-7006.2012.02279.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Revised: 03/07/2012] [Accepted: 03/13/2012] [Indexed: 12/14/2022] Open
Abstract
Recent advances in molecular biology have led to the identification of new molecular targets, such as epidermal growth factor receptor ( EGFR ) mutations and echinoderm microtubule-associated protein-like 4 (EML4) - anaplastic lymphoma kinase (ALK) fusion gene, in lung cancer. Dramatic response has been achieved with EGFR inhibitors (gefitinib and erlotinib) and an ALK inhibitor (crizotinib) in lung cancer expressing corresponding targets. However, cancer cells acquire resistance to these drugs and cause recurrence. Known major mechanisms for resistance to molecular targeted drugs include gatekeeper mutations in the target gene and activation of bypass survival signal via receptors other than the target receptors. The latter mechanism can involve receptor gene amplification and ligand-triggered receptor activation as well. For example, hepatocyte growth factor (HGF), the ligand of a tyrosine kinase receptor Met, activates Met and the downstream PI3K/Akt pathway and triggers resistance to EGFR inhibitors in EGFR mutant lung cancer cells. Moreover, EGFR ligands activate EGFR and downstream pathways and trigger resistance to crizotinib in EML4-ALK lung cancer cells. These observations indicate that signals from oncogenic drivers (EGFR signaling in EGFR -mutant lung cancer and ALK signaling in EML4-ALK lung cancer) and ligand-triggered bypass signals (HGF-Met and EGFR ligands-EGFR, respectively) must be simultaneously blocked to avoid the resistance. This review focuses specifically on receptor activation by ligand stimulation and discusses novel therapeutic strategies that are under development for overcoming resistance to molecular targeted drugs in lung cancer.
Collapse
Affiliation(s)
- Seiji Yano
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Ishikawa.
| | | | | | | |
Collapse
|
138
|
Abstract
The MET pathway is dysregulated in many human cancers and promotes tumour growth, invasion and dissemination. Abnormalities in MET signalling have been reported to correlate with poor clinical outcomes and drug resistance in patients with cancer. Thus, MET has emerged as an attractive target for cancer therapy. Several MET inhibitors have been introduced into the clinic, and are currently in all phases of clinical trials. In general, initial results from these studies indicate only a modest benefit in unselected populations. In this Review, we discuss current challenges in developing MET inhibitors--including identification of predictive biomarkers--as well as the most-efficient ways to combine these drugs with other targeted agents or with classic chemotherapy or radiotherapy.
Collapse
|
139
|
Greenall SA, Gherardi E, Liu Z, Donoghue JF, Vitali AA, Li Q, Murphy R, Iamele L, Scott AM, Johns TG. Non-agonistic bivalent antibodies that promote c-MET degradation and inhibit tumor growth and others specific for tumor related c-MET. PLoS One 2012; 7:e34658. [PMID: 22511956 PMCID: PMC3325269 DOI: 10.1371/journal.pone.0034658] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 03/06/2012] [Indexed: 11/18/2022] Open
Abstract
The c-MET receptor has a function in many human cancers and is a proven therapeutic target. Generating antagonistic or therapeutic monoclonal antibodies (mAbs) targeting c-MET has been difficult because bivalent, intact anti-Met antibodies frequently display agonistic activity, necessitating the use of monovalent antibody fragments for therapy. By using a novel strategy that included immunizing with cells expressing c-MET, we obtained a range of mAbs. These c-MET mAbs were tested for binding specificity and anti-tumor activity using a range of cell-based techniques and in silico modeling. The LMH 80 antibody bound an epitope, contained in the small cysteine-rich domain of c-MET (amino acids 519–561), that was preferentially exposed on the c-MET precursor. Since the c-MET precursor is only expressed on the surface of cancer cells and not normal cells, this antibody is potentially tumor specific. An interesting subset of our antibodies displayed profound activities on c-MET internalization and degradation. LMH 87, an antibody binding the loop connecting strands 3d and 4a of the 7-bladed β-propeller domain of c-MET, displayed no intrinsic agonistic activity but promoted receptor internalization and degradation. LMH 87 inhibited HGF/SF-induced migration of SK-OV-3 ovarian carcinoma cells, the proliferation of A549 lung cancer cells and the growth of human U87MG glioma cells in a mouse xenograft model. These results indicate that c-MET antibodies targeting epitopes controlling receptor internalization and degradation provide new ways of controlling c-MET expression and activity and may enable the therapeutic targeting of c-MET by intact, bivalent antibodies.
Collapse
Affiliation(s)
- Sameer A. Greenall
- Oncogenic Signaling Laboratory, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | | | - Zhanqi Liu
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research, Heidelberg, Victoria, Australia
| | - Jacqueline F. Donoghue
- Oncogenic Signaling Laboratory, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Angela A. Vitali
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research, Heidelberg, Victoria, Australia
| | - Qian Li
- Oncogenic Signaling Laboratory, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Roger Murphy
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research, Heidelberg, Victoria, Australia
| | - Luisa Iamele
- Medical Research Council Centre, Cambridge, United Kingdom
| | - Andrew M. Scott
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research, Heidelberg, Victoria, Australia
| | - Terrance G. Johns
- Oncogenic Signaling Laboratory, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|
140
|
Klotz M, Schmid E, Steiner-Hahn K, Rose T, Laube J, Roese L, Henderson D, Krahn T, von Ahsen O. Preclinical evaluation of biomarkers for response monitoring to the MET inhibitor BAY-853474. Biomarkers 2012; 17:325-35. [PMID: 22452362 DOI: 10.3109/1354750x.2012.670865] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
CONTEXT The receptor tyrosine kinase MET contributes to a wide range of biological activities, including survival, proliferation, and metastasis, which play an important role in cancer progression. MET is frequently overexpressed or amplified in a range of malignancies. Therefore, MET is an attractive therapeutic target for treatment of cancer. BAY-853474 is a novel specific MET inhibitor highly effective in preclinical tumor models. OBJECTIVE For response monitoring in clinical studies, soluble plasma biomarkers are the most convenient and least invasive choice. Therefore, we sought to identify such biomarkers in xenograft models. RESULTS We show that BAY-853474 reduces the tumor burden in U87MG glioblastoma, NCI-H1993 nonsmall cell lung cancer, and HS746T gastric cancer xenograft models. We demonstrate that the dose dependence is reflected by inhibition of MET phosphorylation and that the soluble plasma biomarkers hepatocyte growth factor, vascular endothelial growth factor, and interleukin-8 as well as the MET-ectodomain can be used to monitor the tumor size and response to treatment. Clinical samples, however, show only moderately elevated levels of these biomarker candidates in cancer patients even with MET amplification. We, therefore, established an immunohistochemistry (IHC) protocol to detect MET phosphorylation that is suitable to monitor the effect of BAY-853474 in tumor biopsies. CONCLUSION IHC-based analysis of target phosphorylation in tumor biopsies is recommended in addition to testing plasma biomarkers for response monitoring.
Collapse
Affiliation(s)
- Monika Klotz
- Global Biomarker Research, Bayer Pharma AG, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Sennino B, Ishiguro-Oonuma T, Wei Y, Naylor RM, Williamson CW, Bhagwandin V, Tabruyn SP, You WK, Chapman HA, Christensen JG, Aftab DT, McDonald DM. Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov 2012; 2:270-87. [PMID: 22585997 DOI: 10.1158/2159-8290.cd-11-0240] [Citation(s) in RCA: 332] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
UNLABELLED Invasion and metastasis increase after the inhibition of VEGF signaling in some preclinical tumor models. In the present study we asked whether selective VEGF inhibition is sufficient to increase invasion and metastasis and whether selective c-Met inhibition is sufficient to block this effect. Treatment of pancreatic neuroendocrine tumors in RIP-Tag2 mice with a neutralizing anti-VEGF antibody reduced tumor burden but increased tumor hypoxia, hypoxia-inducible factor-1α, and c-Met activation and also increased invasion and metastasis. However, invasion and metastasis were reduced by concurrent inhibition of c-Met by PF-04217903 or PF-02341066 (crizotinib). A similar benefit was found in orthotopic Panc-1 pancreatic carcinomas treated with sunitinib plus PF-04217903 and in RIP-Tag2 tumors treated with XL184 (cabozantinib), which simultaneously blocks VEGF and c-Met signaling. These findings document that invasion and metastasis are promoted by selective inhibition of VEGF signaling and can be reduced by the concurrent inhibition of c-Met. SIGNIFICANCE This report examines the mechanism of increased tumor aggressiveness after anti-VEGF therapy and presents evidence for roles of vascular pruning, hypoxia, and c-Met activation. The results show that simultaneous inhibition of c-Met and VEGF signaling not only slows tumor growth but also reduces invasion and metastasis.
Collapse
Affiliation(s)
- Barbara Sennino
- Comprehensive Cancer Center, Department of Anatomy, University of California-San Francisco (UCSF), San Francisco, CA 94143-0452, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Surati M, Patel P, Peterson A, Salgia R. Role of MetMAb (OA-5D5) in c-MET active lung malignancies. Expert Opin Biol Ther 2012; 11:1655-62. [PMID: 22047509 DOI: 10.1517/14712598.2011.626762] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION MetMAb (OA-5D5) is a one-armed monoclonal antibody developed to bind to and inhibit c-MET receptor tyrosine kinase. Though only in early clinical testing, this agent holds great promise in diseases thought to be driven by c-MET activation, as evidenced by the Phase II results in NSCLC where a benefit in overall survival was observed in patients with MET-diagnostic-positive disease. Thus far, both alone and in combination with other targeted agents, this drug has been well tolerated and no new significant safety signals have been identified. AREAS COVERED This review summarizes the structure and function of the c-MET receptor and its ligand hepatic growth factor (HGF), provides an overview of select targeted monotherapies developed to interfere in the MET-HGF signaling pathway, discusses pre-clinical and clinical data surrounding MetMAb, and concludes with an expert opinion regarding this novel agent. EXPERT OPINION MetMAb has been well tolerated and based on Phase II data testing it, in combination with erlotinib in advanced NSCLC, may have a role in improving survival in patients with disease driven by c-MET activation. However, Phase III validation is underway and the results of these studies will help elucidate which patients will benefit most from this novel agent.
Collapse
Affiliation(s)
- Mosmi Surati
- University of Chicago, Pritzker School of Medicine, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
143
|
Iovanna J, Mallmann MC, Gonçalves A, Turrini O, Dagorn JC. Current knowledge on pancreatic cancer. Front Oncol 2012; 2:6. [PMID: 22655256 PMCID: PMC3356035 DOI: 10.3389/fonc.2012.00006] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 01/11/2012] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer death with a median survival of 6 months and a dismal 5-year survival rate of 3-5%. The development and progression of pancreatic cancer are caused by the activation of oncogenes, the inactivation of tumor suppressor genes, and the deregulation of many signaling pathways. Therefore, the strategies targeting these molecules as well as their downstream signaling could be promising for the prevention and treatment of pancreatic cancer. However, although targeted therapies for pancreatic cancer have yielded encouraging results in vitro and in animal models, these findings have not been translated into improved outcomes in clinical trials. This failure is due to an incomplete understanding of the biology of pancreatic cancer and to the selection of poorly efficient or imperfectly targeted agents. In this review, we will critically present the current knowledge regarding the molecular, biochemical, clinical, and therapeutic aspects of pancreatic cancer.
Collapse
Affiliation(s)
- Juan Iovanna
- INSERM U624, Stress Cellulaire, Parc Scientifique et Technologique de LuminyMarseille, France
| | | | - Anthony Gonçalves
- Département d’Oncologie Médicale, Institut Paoli-CalmettesMarseille, France
| | - Olivier Turrini
- Département de Chirurgie Oncologique, Institut Paoli-CalmettesMarseille, France
| | - Jean-Charles Dagorn
- INSERM U624, Stress Cellulaire, Parc Scientifique et Technologique de LuminyMarseille, France
| |
Collapse
|
144
|
Abstract
Uncontrolled cell survival, growth, angiogenesis and metastasis are essential hallmarks of cancer. Genetic and biochemical data have demonstrated that the growth and motility factor hepatocyte growth factor/scatter factor (HGF/SF) and its receptor, the tyrosine kinase MET, have a causal role in all of these processes, thus providing a strong rationale for targeting these molecules in cancer. Parallel progress in understanding the structure and function of HGF/SF, MET and associated signalling components has led to the successful development of blocking antibodies and a large number of small-molecule MET kinase inhibitors. In this Review, we discuss these advances, as well as results from recent clinical studies that demonstrate that inhibiting MET signalling in several types of solid human tumours has major therapeutic value.
Collapse
Affiliation(s)
- Ermanno Gherardi
- Medical Research Council (MRC) Centre, Hills Road, Cambridge CB2 2QH, UK.
| | | | | | | |
Collapse
|
145
|
Chattopadhyay C, Ellerhorst JA, Ekmekcioglu S, Greene VR, Davies MA, Grimm EA. Association of activated c-Met with NRAS-mutated human melanomas. Int J Cancer 2012; 131:E56-65. [PMID: 22020736 DOI: 10.1002/ijc.26487] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 09/19/2011] [Indexed: 11/11/2022]
Abstract
Cutaneous melanomas can be divided into three mutually exclusive genetic subsets: tumors with mutated BRAF, tumors with mutated NRAS and tumors wild type at both loci (wt/wt). Targeted therapy for melanoma has been advancing with agents directed to mutated BRAF, accounting for 50% of melanoma patients. The c-Met pathway is known to play a role in melanoma tumorigenesis and preliminary data from our laboratory suggested that this pathway is preferentially activated in NRAS-mutated tumors. The objective of this study was to test the hypothesis that melanomas carrying the mutated NRAS genotype are uniquely sensitively to c-Met inhibition, thus providing rationale for therapeutic targeting of c-Met in this patient cohort. Using primary human melanomas with known BRAF/NRAS genotypes, we observed greater immunostaining for phosphorylated (activated) c-Met in NRAS-mutated and wt/wt tumors, compared to BRAF-mutated tumors. NRAS-mutated and wt/wt cell lines also demonstrated more robust c-Met activation in response to hepatocyte growth factor (HGF). Knock-down of mutated N-Ras, but not wild type N-Ras, by RNA interference resulted in decreased c-Met phosphorylation. Compared to BRAF mutants, NRAS-mutated melanoma cells were more sensitive to pharmacologic c-Met inhibition in terms of c-Met activation, Akt phosphorylation, tumor cell proliferation, migration and apoptosis. This enhanced sensitivity was observed in wt/wt cells as well, but was a less consistent finding. On the basis of these experimental results, we propose that c-Met inhibition may be a useful therapeutic strategy for melanomas with NRAS mutations, as well as some tumors with a wt/wt genotype.
Collapse
Affiliation(s)
- Chandrani Chattopadhyay
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
146
|
Sattler M, Reddy MM, Hasina R, Gangadhar T, Salgia R. The role of the c-Met pathway in lung cancer and the potential for targeted therapy. Ther Adv Med Oncol 2011; 3:171-84. [PMID: 21904579 DOI: 10.1177/1758834011408636] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Hepatocyte growth factor receptor (HGFR), the product of the MET gene, plays an important role in normal cellular function and oncogenesis. In cancer, HGFR has been implicated in cellular proliferation, cell survival, invasion, cell motility, metastasis and angiogenesis. Activation of HGFR can occur through binding to its ligand, hepatocyte growth factor (HGF), overexpression/amplification, mutation, and/or decreased degradation. Amplification of HGFR can occur de novo or in resistance to therapy. Mutations of HGFR have been described in the tyrosine kinase domain, juxtamembrane domain, or semaphorin domain in a number of tumors. These mutations appear to have gain of function, and also reflect differential sensitivity to therapeutic inhibition. There have been various drugs developed to target HGFR, including antibodies to HGFR/HGF, small-molecule inhibitors against the tyrosine kinase domain of HGFR and downstream targets. Different HGFR inhibitors are currently in clinical trials in lung cancer and a number of solid tumors. Several phase I trials have already been completed, and two specific trials have been reported combining HGFR with epidermal growth factor receptor (EGFR) inhibition in non-small cell lung cancer. In particular, trials involving MetMAb and ARQ197 (tivantinib) have gained interest. Ultimately, as individualized therapies become a reality for cancers, HGFR will be an important molecular target.
Collapse
Affiliation(s)
- Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, and Brigham and Women's Hospital, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
147
|
Padhye SS, Guin S, Yao HP, Zhou YQ, Zhang R, Wang MH. Sustained expression of the RON receptor tyrosine kinase by pancreatic cancer stem cells as a potential targeting moiety for antibody-directed chemotherapeutics. Mol Pharm 2011; 8:2310-9. [PMID: 22014215 DOI: 10.1021/mp200193u] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer stem cells (CSCs) contribute to pancreatic cancer tumorigenesis through tumor initiation, drug resistance, and metastasis. Currently, therapeutics targeting pancreatic CSCs are under intensive investigation. This study tested a novel strategy that utilizes the RON receptor as a drug delivery moiety for increased therapeutic activity against pancreatic CSCs. CD24(+)CD44(+)ESA(+) triple-positive pancreatic CSCs (CSCs(+24/44/ESA)) were obtained from spheroids of pancreatic L3.6pl cancer cells by sequential magnetic cell sorting methods. These cells displayed a spherical growth pattern, expressed the unique self-renewal marker Bmi-1, redifferentiated into an epithelial phenotype, acquired an epithelial to mesenchymal phenotype, and caused tumor formation in animal models. Among several receptor tyrosine kinases examined, RON was highly expressed and sustained by CSCs(+24/44/ESA). This feature provided the cellular basis for validating the therapeutic effectiveness of anti-RON antibody Zt/c9-directing doxorubicin-immunoliposomes (Zt/c9-Dox-IL). Zt/c9-Dox-IL specifically interacted with CSCs(+24/44/ESA) and rapidly caused RON internalization, which led to the uptake of liposome-coated Dox. Moreover, Zt/c9-Dox-IL was effective in reducing viability of L3.6pl cells and CSCs(+24/44/ESA). The IC(50) values between free Dox (62.0 ± 3.1 μM) and Zt/c9-Dox-IL (95.0 ± 6.1 μM) treated CSCs(+24/44/ESA) were at relatively comparable levels. In addition, Zt/c9-Dox-IL in combination with small molecule inhibitors lapatinib, sunitinib, or dasatinib further reduced the viability of CSCs(+24/44/ESA). In conclusion, RON expression by CSCs(+24/44/ESA) is a suitable molecule for the targeted delivery of chemoagents. The anti-RON antibody-directed delivery of chemotherapeutics is effective in reducing viability of pancreatic CSCs.
Collapse
Affiliation(s)
- Snehal S Padhye
- School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | | | | | | | | | | |
Collapse
|
148
|
Giaccone G, Wang Y. Strategies for overcoming resistance to EGFR family tyrosine kinase inhibitors. Cancer Treat Rev 2011; 37:456-64. [PMID: 21367530 PMCID: PMC3139833 DOI: 10.1016/j.ctrv.2011.01.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 01/21/2011] [Indexed: 01/14/2023]
Abstract
The first-generation epidermal growth factor receptor tyrosine kinase inhibitors erlotinib and gefitinib have been incorporated into treatment paradigms for patients with advanced non-small cell lung cancer. These agents are particularly effective in a subset of patients whose tumors harbor activating epidermal growth factor receptor mutations. However, most patients do not respond to these tyrosine kinase inhibitors, and those who do will eventually acquire resistance that typically results from a secondary epidermal growth factor receptor mutation (e.g., T790M), mesenchymal-epithelial transition factor amplification, or activation of other signaling pathways. For patients whose tumors have wild-type epidermal growth factor receptor, there are several known mechanisms of initial resistance (e.g., Kirsten rat sarcoma viral oncogene homolog mutations) but these do not account for all cases, suggesting that unknown mechanisms also contribute. To potentially overcome the issue of resistance, next-generation tyrosine kinase inhibitors are being developed, which irreversibly block multiple epidermal growth factor receptor family members (e.g., afatinib [BIBW 2992] and PF-00299804) and/or vascular endothelial growth factor receptor pathways (e.g., BMS-690514 and XL647). In addition, drugs that block parallel signaling pathways or signaling molecules downstream of the epidermal growth factor receptor, such as the insulin-like growth factor-1 receptor and the mammalian target of rapamycin, are undergoing clinical evaluation. As drug resistance appears to be pleomorphic, combinations of drugs or drugs with multiple targets may be more effective in circumventing resistance.
Collapse
Affiliation(s)
- Giuseppe Giaccone
- Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-1906, United States.
| | | |
Collapse
|
149
|
Wilson TR, Lee DY, Berry L, Shames DS, Settleman J. Neuregulin-1-mediated autocrine signaling underlies sensitivity to HER2 kinase inhibitors in a subset of human cancers. Cancer Cell 2011; 20:158-72. [PMID: 21840482 DOI: 10.1016/j.ccr.2011.07.011] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 06/19/2011] [Accepted: 07/21/2011] [Indexed: 11/16/2022]
Abstract
HER2 kinase inhibitors, such as lapatinib, have demonstrated clinical efficacy in HER2-amplified breast cancers. By profiling nearly 700 human cancer cell lines, we identified a subset of non-HER2 amplified cancer cells with striking sensitivity to HER2 kinase inhibition-particularly from head and neck tumors. These cells were found to depend on a neuregulin-1 (NRG1)-mediated autocrine loop driving HER3 activation, which can be disrupted by lapatinib. Elevated NRG1 expression and activated HER3 are strongly associated with lapatinib sensitivity in vitro, and these biomarkers were enriched in a subset of primary head and neck cancer samples. The findings suggest that patients with NRG1-driven tumors lacking HER2 amplification may derive significant clinical benefit from HER2:HER3-directed therapies.
Collapse
Affiliation(s)
- Timothy R Wilson
- Massachusetts General Hospital Cancer Center and Harvard Medical School. Building 149, 13th Street, Charlestown, Boston, MA 02129, USA
| | | | | | | | | |
Collapse
|
150
|
Palmer TD, Ashby WJ, Lewis JD, Zijlstra A. Targeting tumor cell motility to prevent metastasis. Adv Drug Deliv Rev 2011; 63:568-81. [PMID: 21664937 PMCID: PMC3132821 DOI: 10.1016/j.addr.2011.04.008] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 03/31/2011] [Accepted: 04/05/2011] [Indexed: 01/15/2023]
Abstract
Mortality and morbidity in patients with solid tumors invariably result from the disruption of normal biological function caused by disseminating tumor cells. Tumor cell migration is under intense investigation as the underlying cause of cancer metastasis. The need for tumor cell motility in the progression of metastasis has been established experimentally and is supported empirically by basic and clinical research implicating a large collection of migration-related genes. However, there are few clinical interventions designed to specifically target the motility of tumor cells and adjuvant therapy to specifically prevent cancer cell dissemination is severely limited. In an attempt to define motility targets suitable for treating metastasis, we have parsed the molecular determinants of tumor cell motility into five underlying principles including cell autonomous ability, soluble communication, cell-cell adhesion, cell-matrix adhesion, and integrating these determinants of migration on molecular scaffolds. The current challenge is to implement meaningful and sustainable inhibition of metastasis by developing clinically viable disruption of molecular targets that control these fundamental capabilities.
Collapse
Affiliation(s)
- Trenis D. Palmer
- Department of Pathology, Vanderbilt University, C2104A Medical Center North 1161 21 Ave. S., Nashville TN, 37232
| | - William J. Ashby
- Department of Pathology, Vanderbilt University, C2104A Medical Center North 1161 21 Ave. S., Nashville TN, 37232
| | - John D. Lewis
- London Regional Cancer Program, London Health Science Centre, A4-823 790 Commissioners Rd E London ON, N6A 4L6
| | - Andries Zijlstra
- Department of Pathology, Vanderbilt University, C2104A Medical Center North 1161 21 Ave. S., Nashville TN, 37232
| |
Collapse
|