51
|
The opposing roles of laminin-binding integrins in cancer. Matrix Biol 2017; 57-58:213-243. [DOI: 10.1016/j.matbio.2016.08.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/02/2016] [Accepted: 08/17/2016] [Indexed: 02/06/2023]
|
52
|
Abstract
Malignant mesothelioma is an asbestos-related cancer that occurs most commonly in the pleural space and is incurable. Increasing evidence suggests that aberrant receptor tyrosine kinase (RTK)-directed signalling plays a key role in the pathogenesis of this cancer. In the majority of mesotheliomas, up-regulated expression or signalling by Met, the receptor for hepatocyte growth factor (HGF) can be demonstrated. Following binding of ligand, Met relays signals that promote cell survival, proliferation, movement, invasiveness, branching morphogenesis and angiogenesis. Here we describe the HGF/Met axis and review the mechanisms that lead to the aberrant activation of this signalling system in mesothelioma. We also describe the cross-talk that occurs between HGF/Met and a number of other receptors, ligands and co-receptor systems. The prevalent occurrence of HGF/Met dysregulation in patients with mesothelioma sets the scene for the investigation of pharmaceutical inhibitors of this axis. In light of the inter-relationship between HGF/Met and other ligand receptor, combinatorial targeting strategies may provide opportunities for therapeutic advancement in this challenging tumour.
Collapse
|
53
|
Bleymüller WM, Lämmermann N, Ebbes M, Maynard D, Geerds C, Niemann HH. MET-activating Residues in the B-repeat of the Listeria monocytogenes Invasion Protein InlB. J Biol Chem 2016; 291:25567-25577. [PMID: 27789707 DOI: 10.1074/jbc.m116.746685] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/26/2016] [Indexed: 12/20/2022] Open
Abstract
The facultative intracellular pathogen Listeria monocytogenes causes listeriosis, a rare but life-threatening disease. Host cell entry begins with activation of the human receptor tyrosine kinase MET through the bacterial invasion protein InlB, which contains an internalin domain, a B-repeat, and three GW domains. The internalin domain is known to bind MET, but no interaction partner is known for the B-repeat. Adding the B-repeat to the internalin domain potentiates MET activation and is required to stimulate Madin-Darby canine kidney (MDCK) cell scatter. Therefore, it has been hypothesized that the B-repeat may bind a co-receptor on host cells. To test this hypothesis, we mutated residues that might be important for binding an interaction partner. We identified two adjacent residues in strand β2 of the β-grasp fold whose mutation abrogated induction of MDCK cell scatter. Biophysical analysis indicated that these mutations do not alter protein structure. We then tested these mutants in human HT-29 cells that, in contrast to the MDCK cells, were responsive to the internalin domain alone. These assays revealed a dominant negative effect, reducing the activity of a construct of the internalin domain and mutated B-repeat below that of the individual internalin domain. Phosphorylation assays of MET and its downstream targets AKT and ERK confirmed the dominant negative effect. Attempts to identify a host cell receptor for the B-repeat were not successful. We conclude that there is limited support for a co-receptor hypothesis and instead suggest that the B-repeat contributes to MET activation through low affinity homodimerization.
Collapse
Affiliation(s)
- Willem M Bleymüller
- From the Department of Chemistry, Bielefeld University, 33615 Bielefeld, Germany
| | - Nina Lämmermann
- From the Department of Chemistry, Bielefeld University, 33615 Bielefeld, Germany
| | - Maria Ebbes
- From the Department of Chemistry, Bielefeld University, 33615 Bielefeld, Germany
| | - Daniel Maynard
- From the Department of Chemistry, Bielefeld University, 33615 Bielefeld, Germany
| | - Christina Geerds
- From the Department of Chemistry, Bielefeld University, 33615 Bielefeld, Germany
| | - Hartmut H Niemann
- From the Department of Chemistry, Bielefeld University, 33615 Bielefeld, Germany
| |
Collapse
|
54
|
Hamidi H, Pietilä M, Ivaska J. The complexity of integrins in cancer and new scopes for therapeutic targeting. Br J Cancer 2016; 115:1017-1023. [PMID: 27685444 PMCID: PMC5117799 DOI: 10.1038/bjc.2016.312] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/19/2016] [Accepted: 08/19/2016] [Indexed: 12/24/2022] Open
Abstract
Cancer is a complex disease and progresses within a dynamically evolving extracellular matrix that controls virtually every aspect of the tumour and tumour-associated cells. Interactions with the extracellular microenvironment are predominately mediated by a family of cell-surface transmembrane receptors called integrins. Integrin-matrix engagement leads to the formation of adhesion plaques, consisting of signalling and adaptor proteins, at the plasma membrane that link the extracellular matrix to the regulation of the cell cytoskeleton. In this review, we will highlight exciting data that identify new roles for integrins and integrin-dependent signalling in cancer away from the plasma membrane, discuss the implications of integrin-dependent regulation of Met and ErbB2 growth factor receptors and highlight the role of specific integrins in different stages of cancer development including maintenance of cancer stem cells.
Collapse
Affiliation(s)
- Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku, Tykistökatu 6, Turku 20520, Finland
| | - Mika Pietilä
- Turku Centre for Biotechnology, University of Turku, Tykistökatu 6, Turku 20520, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku, Tykistökatu 6, Turku 20520, Finland.,Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| |
Collapse
|
55
|
Brizzi MP, Pignataro D, Tampellini M, Scagliotti GV, Di Maio M. Systemic treatment of hepatocellular carcinoma: why so many failures in the development of new drugs? Expert Rev Anticancer Ther 2016; 16:1053-62. [PMID: 27548441 DOI: 10.1080/14737140.2016.1227706] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION The increasing knowledge of the genomic landscape of hepatocellular carcinoma (HCC) and the development of molecular targeted therapies are a promising background for increasing the number of effective drugs for HCC patients. In recent years, many new drugs have been tested as an alternative to sorafenib or after sorafenib failure. AREAS COVERED In this review, our aim is to describe the randomized trials recently conducted in HCC patients, in order to understand the main reasons potentially related to the failures of many drugs. In addition, we briefly describe the main ongoing trials, that could potentially change the scenario of HCC treatment in the next years. Expert commentary: Heterogeneity of study populations, lack of understanding of critical drivers of tumor progression, risk of liver toxicity associated with experimental agents, flaws in trial design and marginal antitumoral potency can be considered the main reasons for failure of phase III clinical trials in HCC. Most ongoing trials are conducted without any molecular selection criteria, although many drugs could be probably better tested in a molecularly selected population. The knowledge of potential predictive factors for drug efficacy in patients with advanced HCC could improve the chance of obtaining positive results in clinical trials.
Collapse
Affiliation(s)
- Maria Pia Brizzi
- a Division of Medical Oncology, Department of Oncology , University of Turin, San Luigi Gonzaga Hospital , Turin , Italy
| | - Daniele Pignataro
- a Division of Medical Oncology, Department of Oncology , University of Turin, San Luigi Gonzaga Hospital , Turin , Italy
| | - Marco Tampellini
- a Division of Medical Oncology, Department of Oncology , University of Turin, San Luigi Gonzaga Hospital , Turin , Italy
| | - Giorgio Vittorio Scagliotti
- a Division of Medical Oncology, Department of Oncology , University of Turin, San Luigi Gonzaga Hospital , Turin , Italy
| | - Massimo Di Maio
- b Division of Medical Oncology, Department of Oncology , University of Turin, Mauriziano Hospital , Turin , Italy
| |
Collapse
|
56
|
Kim JY, Welsh EA, Fang B, Bai Y, Kinose F, Eschrich SA, Koomen JM, Haura EB. Phosphoproteomics Reveals MAPK Inhibitors Enhance MET- and EGFR-Driven AKT Signaling in KRAS-Mutant Lung Cancer. Mol Cancer Res 2016; 14:1019-1029. [PMID: 27422710 DOI: 10.1158/1541-7786.mcr-15-0506] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/30/2016] [Indexed: 12/11/2022]
Abstract
Pathway inhibition of the RAS-driven MAPK pathway using small-molecule kinase inhibitors has been a key focus for treating cancers driven by oncogenic RAS, yet significant clinical responses are lacking. Feedback reactivation of ERK driven by drug-induced RAF activity has been suggested as one of the major drug resistance mechanisms, especially in the context of oncogenic RAS. To determine whether additional adaptive resistance mechanisms may coexist, we characterized global phosphoproteomic changes after MEK inhibitor selumetinib (AZD6244) treatment in KRAS-mutant A427 and A549 lung adenocarcinoma cell lines employing mass spectrometry-based phosphoproteomics. We identified 9,075 quantifiable unique phosphosites (corresponding to 3,346 unique phosphoproteins), of which 567 phosphosites were more abundant and 512 phosphosites were less abundant after MEK inhibition. Selumetinib increased phosphorylation of KSR-1, a scaffolding protein required for assembly of MAPK signaling complex, as well as altered phosphorylation of GEF-H1, a novel regulator of KSR-1 and implicated in RAS-driven MAPK activation. Moreover, selumetinib reduced inhibitory serine phosphorylation of MET at Ser985 and potentiated HGF- and EGF-induced AKT phosphorylation. These results were recapitulated by pan-RAF (LY3009120), MEK (GDC0623), and ERK (SCH772984) inhibitors, which are currently under early-phase clinical development against RAS-mutant cancers. Our results highlight the unique adaptive changes in MAPK scaffolding proteins (KSR-1, GEF-H1) and in RTK signaling, leading to enhanced PI3K-AKT signaling when the MAPK pathway is inhibited. IMPLICATIONS This study highlights the unique adaptive changes in MAPK scaffolding proteins (KSR-1, GEF-H1) and in RTK signaling, leading to enhanced PI3K/AKT signaling when the MAPK pathway is inhibited. Mol Cancer Res; 14(10); 1019-29. ©2016 AACR.
Collapse
Affiliation(s)
- Jae-Young Kim
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Eric A Welsh
- Cancer Informatics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Bin Fang
- Proteomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Yun Bai
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Fumi Kinose
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Steven A Eschrich
- Department of Bioinformatics & Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - John M Koomen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| |
Collapse
|
57
|
Barrow-McGee R, Kishi N, Joffre C, Ménard L, Hervieu A, Bakhouche BA, Noval AJ, Mai A, Guzmán C, Robert-Masson L, Iturrioz X, Hulit J, Brennan CH, Hart IR, Parker PJ, Ivaska J, Kermorgant S. Beta 1-integrin-c-Met cooperation reveals an inside-in survival signalling on autophagy-related endomembranes. Nat Commun 2016; 7:11942. [PMID: 27336951 PMCID: PMC4931016 DOI: 10.1038/ncomms11942] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/13/2016] [Indexed: 12/20/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) and integrins cooperate to stimulate cell migration and tumour metastasis. Here we report that an integrin influences signalling of an RTK, c-Met, from inside the cell, to promote anchorage-independent cell survival. Thus, c-Met and β1-integrin co-internalize and become progressively recruited on LC3B-positive 'autophagy-related endomembranes' (ARE). In cells growing in suspension, β1-integrin promotes sustained c-Met-dependent ERK1/2 phosphorylation on ARE. This signalling is dependent on ATG5 and Beclin1 but not on ATG13, suggesting ARE belong to a non-canonical autophagy pathway. This β1-integrin-dependent c-Met-sustained signalling on ARE supports anchorage-independent cell survival and growth, tumorigenesis, invasion and lung colonization in vivo. RTK-integrin cooperation has been assumed to occur at the plasma membrane requiring integrin 'inside-out' or 'outside-in' signalling. Our results report a novel mode of integrin-RTK cooperation, which we term 'inside-in signalling'. Targeting integrin signalling in addition to adhesion may have relevance for cancer therapy.
Collapse
Affiliation(s)
- Rachel Barrow-McGee
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Naoki Kishi
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Carine Joffre
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Ludovic Ménard
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Alexia Hervieu
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Bakhouche A. Bakhouche
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Alejandro J. Noval
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Anja Mai
- University of Turku, Centre for Biotechnology and VTT Technical Research Centre of Finland, FI-20520 Turku, Finland
| | - Camilo Guzmán
- University of Turku, Centre for Biotechnology and VTT Technical Research Centre of Finland, FI-20520 Turku, Finland
| | - Luisa Robert-Masson
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Xavier Iturrioz
- Protein Phosphorylation Laboratory, Francis Crick Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, UK
| | - James Hulit
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Caroline H. Brennan
- School of Biological and Chemical Sciences, Queen Mary University of London, 327 Mile End Road, London E1 4NS, UK
| | - Ian R. Hart
- Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Peter J. Parker
- Protein Phosphorylation Laboratory, Francis Crick Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, UK
- Division of Cancer Studies, King's College School of Medicine, St Thomas Street, London SE1 1UL, UK
| | - Johanna Ivaska
- University of Turku, Centre for Biotechnology and VTT Technical Research Centre of Finland, FI-20520 Turku, Finland
- Department of Biochemistry and Food Chemistry, University of Turku, FI-20520 Turku, Finland
| | - Stéphanie Kermorgant
- Spatial Signalling Team, Centre for Tumour Biology, Barts Cancer Institute—A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
58
|
Ilangumaran S, Villalobos-Hernandez A, Bobbala D, Ramanathan S. The hepatocyte growth factor (HGF)–MET receptor tyrosine kinase signaling pathway: Diverse roles in modulating immune cell functions. Cytokine 2016; 82:125-39. [PMID: 26822708 DOI: 10.1016/j.cyto.2015.12.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/11/2015] [Accepted: 12/12/2015] [Indexed: 12/14/2022]
|
59
|
Jacków J, Löffek S, Nyström A, Bruckner-Tuderman L, Franzke CW. Collagen XVII Shedding Suppresses Re-Epithelialization by Directing Keratinocyte Migration and Dampening mTOR Signaling. J Invest Dermatol 2016; 136:1031-1041. [PMID: 26827763 DOI: 10.1016/j.jid.2016.01.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/10/2015] [Accepted: 01/05/2016] [Indexed: 12/16/2022]
Abstract
Transmembrane collagen XVII is traditionally viewed as an important hemidesmosomal attachment component that promotes stable dermal-epidermal adhesion in the skin. However, its expression is highly elevated at the leading edges of cutaneous wounds or invasive carcinomas, suggesting alternative functions in cell migration. The collagenous ectodomain of collagen XVII is constitutively shed from the cell surface by a disintegrin and metalloproteinases, and this shedding is strongly induced during wound healing. Recently, we investigated the physiological relevance of collagen XVII shedding by generating knock-in mice, which exclusively express a functional non-sheddable collagen XVII mutant. Prevention of ectodomain shedding in these mice caused no spontaneous phenotype in resting skin, but accelerated re-epithelialization on skin wounding. Here, we investigated the mechanistic function of shedding during wound healing. Using the non-shedding collagen XVII mice as a model, we uncovered ectodomain shedding as a highly dynamic modulator of in vivo proliferation and motility of activated keratinocytes through tight coordination of α6β4 integrin-laminin-332 interactions and dampening of mechanistic target of rapamycin signaling at the wound edges. Thus, our studies identify ectodomain shedding of collagen XVII as an interactive platform that translates shedding into a signal for directed cell growth and motility during skin regeneration.
Collapse
Affiliation(s)
- Joanna Jacków
- Department of Dermatology, Medical Center, University of Freiburg, Germany.
| | - Stefanie Löffek
- Department of Dermatology, Medical Center, University of Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Medical Center, University of Freiburg, Germany
| | | | | |
Collapse
|
60
|
Atanelishvili I, Shirai Y, Akter T, Buckner T, Noguchi A, Silver RM, Bogatkevich GS. M10, a caspase cleavage product of the hepatocyte growth factor receptor, interacts with Smad2 and demonstrates antifibrotic properties in vitro and in vivo. Transl Res 2016; 170:99-111. [PMID: 26772959 PMCID: PMC4789156 DOI: 10.1016/j.trsl.2015.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/13/2015] [Accepted: 12/16/2015] [Indexed: 01/07/2023]
Abstract
Hepatocyte growth factor receptor, also known as cellular mesenchymal-epithelial transition factor (c-MET, MET), is an important antifibrotic molecule that protects various tissues, including lung, from injury and fibrosis. The intracellular cytoplasmic tail of MET contains a caspase-3 recognition motif "DEVD-T" that on cleavage by caspase-3 generates a 10-amino acid peptide, TRPASFWETS, designated as "M10". M10 contains at its N-terminus the uncharged amino acid proline (P) directly after a cationic amino acid arginine (R) which favors the transport of the peptide through membranes. M10, when added to cell culture medium, remains in the cytoplasm and nuclei of cells for up to 24 hours. M10 effectively decreases collagen in both scleroderma and TGFβ-stimulated normal lung and skin fibroblasts. M10 interacts with the Mad Homology 2 domain of Smad2 and inhibits TGFβ-induced Smad2 phosphorylation, suggesting that the antifibrotic effects of M10 are mediated in part by counteracting Smad-dependent fibrogenic pathways. In the bleomycin murine model of pulmonary fibrosis, M10 noticeably reduced lung inflammation and fibrosis. Ashcroft fibrosis scores and lung collagen content were significantly lower in bleomycin-treated mice receiving M10 as compared with bleomycin-treated mice receiving scrambled peptide. We conclude that M10 peptide interacts with Smad2 and demonstrates strong antifibrotic effects in vitro and in vivo in an animal model of lung fibrosis and should be considered as a potential therapeutic agent for systemic sclerosis and other fibrosing diseases.
Collapse
Affiliation(s)
- Ilia Atanelishvili
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Yuichiro Shirai
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; Department of Allergy and Rheumatology, Nippon Medical School, Tokyo, Japan
| | - Tanjina Akter
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Taylor Buckner
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; South Carolina Governor's School for Science & Mathematics; Honors College at the College of Charleston, USA
| | - Atsushi Noguchi
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Richard M Silver
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Galina S Bogatkevich
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
61
|
Finisguerra V, Prenen H, Mazzone M. Preclinical and clinical evaluation of MET functions in cancer cells and in the tumor stroma. Oncogene 2016; 35:5457-5467. [PMID: 26996670 DOI: 10.1038/onc.2016.36] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/09/2016] [Accepted: 01/09/2016] [Indexed: 02/06/2023]
Abstract
A lot of attention has been dedicated to investigate the role of the tyrosine kinase receptor MET in tumors. The acquired notion that cancer cells from different histological origin strictly rely on the engagement of this specific oncogene for their growth and survival has certainly justified the development and the use of MET-targeted therapies in the clinic. However, the function and involvement of this pathway in the stroma (that often constitutes >50% of the global cellularity of the tumor) may offer the opportunity to conceive new patient stratification criteria, rational drug design and guided trials of new combination treatments. In this review, we will summarize and discuss the role of MET in cancer cells but especially in different stromal compartments, in light of the results showed by past and recent preclinical and clinical trials with anti-MET drugs.
Collapse
Affiliation(s)
- V Finisguerra
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - H Prenen
- Digestive Oncology, University Hospitals Leuven and Department of Oncology, KU Leuven, Leuven, Belgium
| | - M Mazzone
- Lab of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium.,Lab of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
62
|
Okuma HS, Kondo S. Trends in the development of MET inhibitors for hepatocellular carcinoma. Future Oncol 2016; 12:1275-86. [PMID: 26984595 DOI: 10.2217/fon.16.3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Hepatocellular carcinoma is the third most common cause of cancer-related deaths worldwide. The multikinase inhibitor sorafenib has improved survival and is now considered the standard of care; however, the benefits are still disappointing, and thus, new effective treatments are required. In human hepatocellular carcinoma, MET, which is encoded by the HGFR gene, is activated by amplification, overexpression or mutation, and it has recently emerged as a possible therapeutic target in various tumors including hepatocellular carcinoma. In fact, some drugs targeting the HGF/MET axis are currently under investigation in clinical trials. Here, we review the role of MET and trends in the development of MET inhibitors for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hitomi S Okuma
- Department of Experimental Therapeutics, National Cancer Center, Tokyo, Japan
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, National Cancer Center, Tokyo, Japan
| |
Collapse
|
63
|
Carpenter BL, Chen M, Knifley T, Davis KA, Harrison SMW, Stewart RL, O'Connor KL. Integrin α6β4 Promotes Autocrine Epidermal Growth Factor Receptor (EGFR) Signaling to Stimulate Migration and Invasion toward Hepatocyte Growth Factor (HGF). J Biol Chem 2015; 290:27228-27238. [PMID: 26381405 DOI: 10.1074/jbc.m115.686873] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Indexed: 12/14/2022] Open
Abstract
Integrin α6β4 is up-regulated in pancreatic adenocarcinomas where it contributes to carcinoma cell invasion by altering the transcriptome. In this study, we found that integrin α6β4 up-regulates several genes in the epidermal growth factor receptor (EGFR) pathway, including amphiregulin (AREG), epiregulin (EREG), and ectodomain cleavage protease MMP1, which is mediated by promoter demethylation and NFAT5. The correlation of these genes with integrin α6β4 was confirmed in The Cancer Genome Atlas Pancreatic Cancer Database. Based on previous observations that integrin α6β4 cooperates with c-Met in pancreatic cancers, we examined the impact of EGFR signaling on hepatocyte growth factor (HGF)-stimulated migration and invasion. We found that AREG and EREG were required for autocrine EGFR signaling, as knocking down either ligand inhibited HGF-mediated migration and invasion. We further determined that HGF induced secretion of AREG, which is dependent on integrin-growth factor signaling pathways, including MAPK, PI3K, and PKC. Moreover, matrix metalloproteinase activity and integrin α6β4 signaling were required for AREG secretion. Blocking EGFR signaling with EGFR-specific antibodies or an EGFR tyrosine kinase inhibitor hindered HGF-stimulated pancreatic carcinoma cell chemotaxis and invasive growth in three-dimensional culture. Finally, we found that EGFR was phosphorylated in response to HGF stimulation that is dependent on EGFR kinase activity; however, c-Met phosphorylation in response to HGF was unaffected by EGFR signaling. Taken together, these data illustrate that integrin α6β4 stimulates invasion by promoting autocrine EGFR signaling through transcriptional up-regulation of key EGFR family members and by facilitating HGF-stimulated EGFR ligand secretion. These signaling events, in turn, promote pancreatic carcinoma migration and invasion.
Collapse
Affiliation(s)
- Brittany L Carpenter
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40506-0509; Departments of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40506-0509
| | - Min Chen
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40506-0509; Departments of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40506-0509
| | - Teresa Knifley
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40506-0509
| | - Kelley A Davis
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40506-0509
| | - Susan M W Harrison
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40506-0509
| | - Rachel L Stewart
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40506-0509; Departments of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky 40506-0509
| | - Kathleen L O'Connor
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40506-0509; Departments of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40506-0509.
| |
Collapse
|
64
|
Wang H, Jin H, Rapraeger AC. Syndecan-1 and Syndecan-4 Capture Epidermal Growth Factor Receptor Family Members and the α3β1 Integrin Via Binding Sites in Their Ectodomains: NOVEL SYNSTATINS PREVENT KINASE CAPTURE AND INHIBIT α6β4-INTEGRIN-DEPENDENT EPITHELIAL CELL MOTILITY. J Biol Chem 2015; 290:26103-13. [PMID: 26350464 DOI: 10.1074/jbc.m115.679084] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Indexed: 11/06/2022] Open
Abstract
The α6β4 integrin is known to associate with receptor tyrosine kinases when engaged in epithelial wound healing and in carcinoma invasion and survival. Prior work has shown that HER2 associates with α6β4 integrin and syndecan-1 (Sdc1), in which Sdc1 engages the cytoplasmic domain of the β4 integrin subunit allowing HER2-dependent motility and carcinoma cell survival. In contrast, EGFR associates with Sdc4 and the α6β4 integrin, and EGFR-dependent motility depends on cytoplasmic engagement of β4 integrin with Sdc4. However, how HER2 and EGFR assimilate into a complex with the syndecans and integrin, and why kinase capture is syndecan-specific has remained unknown. In the present study, we demonstrate that HER2 is captured via a site, comprised of amino acids 210-240, in the extracellular domain of human Sdc1, and EGFR is captured via an extracellular site comprised of amino acids 87-131 in human Sdc4. Binding assays using purified recombinant proteins demonstrate that the interaction between the EGFR family members and the syndecans is direct. The α3β1 integrin, which is responsible for the motility of the cells, is captured at these sites as well. Peptides based on the interaction motifs in Sdc1 and Sdc4, called synstatins (SSTN210-240 and SSTN87-131) competitively displace the receptor tyrosine kinase and α3β1 integrin from the syndecan with an IC50 of 100-300 nm. The syndecans remain anchored to the α6β4 integrin via its cytoplasmic domain, but the activation of cell motility is disrupted. These novel SSTN peptides are potential therapeutics for carcinomas that depend on these HER2- and EGFR-coupled mechanisms for their invasion and survival.
Collapse
Affiliation(s)
| | | | - Alan C Rapraeger
- From the Department of Human Oncology and the University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, Wisconsin 53705
| |
Collapse
|
65
|
Clinical significance of the integrin α6β4 in human malignancies. J Transl Med 2015; 95:976-86. [PMID: 26121317 PMCID: PMC4554527 DOI: 10.1038/labinvest.2015.82] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 05/06/2015] [Accepted: 05/13/2015] [Indexed: 12/14/2022] Open
Abstract
Integrin α6β4 is a cellular adhesion molecule that binds to laminins in the extracellular matrix and nucleates the formation of hemidesmosomes. During carcinoma progression, integrin α6β4 is released from hemidesmosomes, where it can then signal to facilitate multiple aspects of tumor progression including sustaining proliferative signaling, tumor invasion and metastasis, evasion of apoptosis, and stimulation of angiogenesis. The integrin achieves these ends by cooperating with growth factor receptors including EGFR, ErbB-2, and c-Met to amplify downstream pathways such as PI3K, AKT, MAPK, and the Rho family small GTPases. Furthermore, it dramatically alters the transcriptome toward a more invasive phenotype by controlling promoter DNA demethylation of invasion and metastasis-associated proteins, such as S100A4 and autotaxin, and upregulates and activates key tumor-promoting transcription factors such as the NFATs and NF-κB. Expression of integrin α6β4 has been studied in many human malignancies where its overexpression is associated with aggressive behavior and a poor prognosis. This review provides an assessment of integrin α6β4 expression patterns and their prognostic significance in human malignancies, and describes key signaling functions of integrin α6β4 that contribute to tumor progression.
Collapse
|
66
|
Marano L, Chiari R, Fabozzi A, De Vita F, Boccardi V, Roviello G, Petrioli R, Marrelli D, Roviello F, Patriti A. c-Met targeting in advanced gastric cancer: An open challenge. Cancer Lett 2015; 365:30-36. [PMID: 26049023 DOI: 10.1016/j.canlet.2015.05.028] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 05/21/2015] [Accepted: 05/23/2015] [Indexed: 12/14/2022]
Abstract
Despite significant improvements in systemic chemotherapy over the last two decades, the prognosis of patients with advanced gastric and gastroesophageal junction adenocarcinoma (GC) remains poor. Because of molecular heterogeneity, it is essential to classify tumors based on the underlying oncogenic pathways and to develop targeted therapies acting on individual tumors. High-quality research and advances in technology have contributed to the elucidation of molecular pathways underlying disease progression and have stimulated many clinical studies testing target therapies in an advanced disease setting. In particular, strong preclinical evidence for the aberrant activation of the HGF/c-Met signaling pathways in GC cancers exists. This review will cover the c-Met pathway, the mechanisms of c-Met activation and the different strategies of its inhibition. Next, we will focus on the current state of the art in the clinical evaluation of c-Met-targeted therapies and the description of ongoing randomized trials with the idea that in this disease, high quality translational research to identify and validate biomarkers is a priority task.
Collapse
Affiliation(s)
- Luigi Marano
- General, Minimally Invasive and Robotic Surgery, Department of Surgery, "San Matteo degli Infermi" Hospital, ASL Umbria 2, 06049 Spoleto, Italy.
| | - Rita Chiari
- Department of Medical Oncology, "Santa Maria della Misericordia" Hospital, Azienda Ospedaliera di Perugia, 06132 Perugia, Italy
| | - Alessio Fabozzi
- Division of Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi-A. Lanzara", Second University of Naples, 80131 Naples, Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi-A. Lanzara", Second University of Naples, 80131 Naples, Italy
| | - Virginia Boccardi
- Section of Gerontology and Geriatrics, Department of Medicine, University of Perugia, 06132 Perugia, Italy
| | | | | | - Daniele Marrelli
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Franco Roviello
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Alberto Patriti
- General, Minimally Invasive and Robotic Surgery, Department of Surgery, "San Matteo degli Infermi" Hospital, ASL Umbria 2, 06049 Spoleto, Italy
| |
Collapse
|
67
|
Shi R, Li J, Tang F, Luo YI, Tu CQ. Identification and functional study of osteosarcoma metastasis marker genes. Oncol Lett 2015; 10:1848-1852. [PMID: 26622763 DOI: 10.3892/ol.2015.3444] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 05/07/2015] [Indexed: 02/05/2023] Open
Abstract
To date, osteosarcoma metastasis genes, which are key for accurate initial diagnosis of the disease, have not been well identified. In the present study, osteosarcoma samples with and without metastasis were collected from 31 patients. Specific complementary DNA subtraction techniques were used to identify the osteosarcoma metastasis transcripts, which are responsible for the metastasis of osteosarcoma. The specific differentially expressed transcripts were identified by Basic Local Alignment Search Tool analysis and the results were validated by immunoblotting. Specifically, ezrin and β4 integrin were employed as markers to detect osteosarcoma metastasis in the initial stages. The results of the present study indicated that the two transcripts, ezrin and β4 integrin, were highly expressed in patients with osteosarcoma metastasis, and concluded that these were osteosarcoma metastasis genes. These results indicate that β4 integrin and/or ezrin may be used as a novel marker for the detection of osteosarcoma metastasis in the initial stages.
Collapse
Affiliation(s)
- Rui Shi
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Juan Li
- Department of Pulmonary Tumors, Sichuan Cancer Hospital, Chengdu, Sichuan 610041, P.R. China
| | - Fan Tang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Y I Luo
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chong-Qi Tu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
68
|
Hepatocyte Growth Factor Receptor c-Met Instructs T Cell Cardiotropism and Promotes T Cell Migration to the Heart via Autocrine Chemokine Release. Immunity 2015; 42:1087-99. [PMID: 26070483 PMCID: PMC4510150 DOI: 10.1016/j.immuni.2015.05.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 10/18/2014] [Accepted: 04/02/2015] [Indexed: 12/20/2022]
Abstract
Effector-T-cell-mediated immunity depends on the efficient localization of antigen-primed lymphocytes to antigen-rich non-lymphoid tissue, which is facilitated by the expression of a unique set of “homing” receptors acquired by memory T cells. We report that engagement of the hepatocyte growth factor (HGF) receptor c-Met by heart-produced HGF during priming in the lymph nodes instructs T cell cardiotropism, which was associated with a specialized homing “signature” (c-Met+CCR4+CXCR3+). c-Met signals facilitated T cell recruitment to the heart via the chemokine receptor CCR5 by inducing autocrine CCR5 ligand release. c-Met triggering was sufficient to support cardiotropic T cell recirculation, while CCR4 and CXCR3 sustained recruitment during heart inflammation. Transient pharmacological blockade of c-Met during T cell priming led to enhanced survival of heart, but not skin, allografts associated with impaired localization of alloreactive T cells to heart grafts. These findings suggest c-Met as a target for development of organ-selective immunosuppressive therapies. HGF-induced c-Met signals during activation induce cardiotropic memory T cells Cardiotropic T cells express a specific molecular signature (c-Met+CCR4+CXCR3+) By inducing an autocrine chemokine loop, c-Met also promotes T cell recruitment Blockade of the HGF-c-Met axis prevents heart, but not skin, allograft rejection
Collapse
|
69
|
Molecular architecture and function of the hemidesmosome. Cell Tissue Res 2015; 360:529-44. [PMID: 26017636 PMCID: PMC4452579 DOI: 10.1007/s00441-015-2216-6] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 11/03/2014] [Indexed: 01/13/2023]
Abstract
Hemidesmosomes are multiprotein complexes that facilitate the stable adhesion of basal epithelial cells to the underlying basement membrane. The mechanical stability of hemidesmosomes relies on multiple interactions of a few protein components that form a membrane-embedded tightly-ordered complex. The core of this complex is provided by integrin α6β4 and P1a, an isoform of the cytoskeletal linker protein plectin that is specifically associated with hemidesmosomes. Integrin α6β4 binds to the extracellular matrix protein laminin-332, whereas P1a forms a bridge to the cytoplasmic keratin intermediate filament network. Other important components are BPAG1e, the epithelial isoform of bullous pemphigoid antigen 1, BPAG2, a collagen-type transmembrane protein and CD151. Inherited or acquired diseases in which essential components of the hemidesmosome are missing or structurally altered result in tissue fragility and blistering. Modulation of hemidesmosome function is of crucial importance for a variety of biological processes, such as terminal differentiation of basal keratinocytes and keratinocyte migration during wound healing and carcinoma invasion. Here, we review the molecular characteristics of the proteins that make up the hemidesmosome core structure and summarize the current knowledge about how their assembly and turnover are regulated by transcriptional and post-translational mechanisms.
Collapse
|
70
|
Petrini I. Biology of MET: a double life between normal tissue repair and tumor progression. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:82. [PMID: 25992381 DOI: 10.3978/j.issn.2305-5839.2015.03.58] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 01/28/2015] [Indexed: 01/30/2023]
Abstract
MNNG HOS transforming gene (MET) is a class IV receptor tyrosine kinase, expressed on the surface of epithelial cells. The interaction with the hepatocyte grow factor (HGF) induces MET dimerization and the activation of multiple intracellular pathways leading to cell proliferation, anti-apoptosis, morphogenic differentiation, motility, invasion, and angiogenesis. Knock out mice have demonstrated that MET is necessary for normal embryogenesis including the formation of striate muscles, liver and trophoblastic structures. The overexpression of MET and HGF are common in solid tumors and contribute to determine their growth. Indeed, MET has been cloned as a transforming gene from a chemically induced human osteosarcoma cell line and therefore is considered a proto-oncogene. Germline MET mutations are characteristic of hereditary papillary kidney cancers and MET amplification is observed in tumors including lung and gastric adenocarcinomas. The inhibition of MET signaling is the target for specific drugs that are raising exciting expectation for medical treatment of cancer.
Collapse
|
71
|
Ascolani G, Occhipinti A, Liò P. Modelling circulating tumour cells for personalised survival prediction in metastatic breast cancer. PLoS Comput Biol 2015; 11:e1004199. [PMID: 25978366 PMCID: PMC4433130 DOI: 10.1371/journal.pcbi.1004199] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 02/16/2015] [Indexed: 12/16/2022] Open
Abstract
Ductal carcinoma is one of the most common cancers among women, and the main cause of death is the formation of metastases. The development of metastases is caused by cancer cells that migrate from the primary tumour site (the mammary duct) through the blood vessels and extravasating they initiate metastasis. Here, we propose a multi-compartment model which mimics the dynamics of tumoural cells in the mammary duct, in the circulatory system and in the bone. Through a branching process model, we describe the relation between the survival times and the four markers mainly involved in metastatic breast cancer (EPCAM, CD47, CD44 and MET). In particular, the model takes into account the gene expression profile of circulating tumour cells to predict personalised survival probability. We also include the administration of drugs as bisphosphonates, which reduce the formation of circulating tumour cells and their survival in the blood vessels, in order to analyse the dynamic changes induced by the therapy. We analyse the effects of circulating tumour cells on the progression of the disease providing a quantitative measure of the cell driver mutations needed for invading the bone tissue. Our model allows to design intervention scenarios that alter the patient-specific survival probability by modifying the populations of circulating tumour cells and it could be extended to other cancer metastasis dynamics.
Collapse
Affiliation(s)
- Gianluca Ascolani
- University of Cambridge, Computer Laboratory, Cambridge, United Kingdom
| | | | - Pietro Liò
- University of Cambridge, Computer Laboratory, Cambridge, United Kingdom
| |
Collapse
|
72
|
Granito A, Guidetti E, Gramantieri L. c-MET receptor tyrosine kinase as a molecular target in advanced hepatocellular carcinoma. J Hepatocell Carcinoma 2015; 2:29-38. [PMID: 27508192 PMCID: PMC4918282 DOI: 10.2147/jhc.s77038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
c-MET is the membrane receptor for hepatocyte growth factor (HGF), also known as scatter factor or tumor cytotoxic factor, a mitogenic growth factor for hepatocytes. HGF is mainly produced by cells of mesenchymal origin and it mainly acts on neighboring epidermal and endothelial cells, regulating epithelial growth and morphogenesis. HGF/MET signaling has been identified among the drivers of tumorigenesis in human cancers. As such, c-MET is a recognized druggable target, and against it, targeted agents are currently under clinical investigation. c-MET overexpression is a common event in a wide range of human malignancies, including gastric, lung, breast, ovary, colon, kidney, thyroid, and liver carcinomas. Despite c-MET overexpression being reported by a large majority of studies, no evidence for a c-MET oncogenic addiction exists in hepatocellular carcinoma (HCC). In particular, c-MET amplification is a rare event, accounting for 4%–5% of cases while no mutation has been identified in c-MET oncogene in HCC. Thus, the selection of patient subgroups more likely to benefit from c-MET inhibition is challenging. Notwithstanding, c-MET overexpression was reported to be associated with increased metastatic potential and poor prognosis in patients with HCC, providing a rationale for its therapeutic inhibition. Here we summarize the role of activated HGF/MET signaling in HCC, its prognostic relevance, and the implications for therapeutic approaches in HCC.
Collapse
Affiliation(s)
- Alessandro Granito
- Dipartimento di Scienze Mediche e Chirurgiche Università di Bologna, Bologna, Italy
| | - Elena Guidetti
- Dipartimento di Scienze Mediche e Chirurgiche Università di Bologna, Bologna, Italy
| | - Laura Gramantieri
- Dipartimento dell'Apparato Digerente, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Centro di Ricerca Biomedica Applicata (CRBA), Azienda Ospedaliero-Universitaria Policlinico S Orsola-Malpighi e Università di Bologna, Bologna, Italy
| |
Collapse
|
73
|
Zorzetto M, Ferrari S, Saracino L, Inghilleri S, Stella GM. MET genetic lesions in non-small-cell lung cancer: pharmacological and clinical implications. Transl Lung Cancer Res 2015; 1:194-207. [PMID: 25806181 DOI: 10.3978/j.issn.2218-6751.2012.09.03] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 09/14/2012] [Indexed: 12/14/2022]
Abstract
Lung cancer is the leading cause of death for solid tumors worldwide with an annual mortality of over one million. Lung carcinoma includes a series of different diseases which are roughly divided into two groups based on clinical and histo-pathological features: non-small cell lung cancer (NSCLC), accounting for almost 80% of lung cancer diagnosis and small cell lung cancer (SCLC) responsible for the remaining 20%. The NSCLC molecular profile has been deeply investigated; alterations in several oncogenes, tumor suppressor genes and transcription factors have been detected, mainly in adenocarcinomas. Dissection of such a complex scenario represents a still open challenge for both researchers and clinicians. MET, the receptor for Hepatocyte Growth Factor (HGF), has been recently identified as a novel promising target in several human malignancies, including NSCLC. Deregulation of the HGF/MET signaling pathway can occur via different mechanisms, including HGF and/or MET overexpression, MET gene amplification, mutations or rearrangements. While the role of MET mutations in NSCLC is not yet fully understood, MET amplification emerged as a critical event in driving cell survival, with preclinical data suggesting that MET-amplified cell lines are exquisitely sensitive to MET inhibition. True MET amplification, which has been associated with poor prognosis in different retrospective series, is a relatively uncommon event in NSCLC, occurring in 1-7% of unselected cases. Nevertheless, in highly selected cohorts of patients, such as those harboring somatic mutations of EGFR with acquired resistance to EGFR tyrosine kinase inhibitors, MET amplification can be observed in up to 20% of cases. Preclinical data suggested that a treatment approach including a combination of EGFR and MET tyrosine kinases could be an effective strategy in this setting and led to the clinical investigation of multiple MET inhibitors in combination with anti-EGFR agents. Results from ongoing and future trials will clarify the role of anti-MET molecules for the treatment of NSCLC and will provide insights into the most appropriate timing for their use. The present review recapitulates the current knowledge on the role of MET signaling in NSCLC mainly focusing on its implications in molecular diagnostic approach and on the novel targeted inhibitors.
Collapse
Affiliation(s)
- Michele Zorzetto
- Department of Molecular Medicine, - Section of Pneumology, Laboratory of Biochemistry & Genetics; University and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia- Italy
| | - Simona Ferrari
- Department of Molecular Medicine, - Section of Pneumology, Laboratory of Biochemistry & Genetics; University and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia- Italy
| | - Laura Saracino
- Department of Molecular Medicine, - Section of Pneumology, Laboratory of Biochemistry & Genetics; University and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia- Italy
| | - Simona Inghilleri
- Department of Molecular Medicine, - Section of Pneumology, Laboratory of Biochemistry & Genetics; University and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia- Italy
| | - Giulia M Stella
- Department of Molecular Medicine, - Section of Pneumology, Laboratory of Biochemistry & Genetics; University and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia- Italy
| |
Collapse
|
74
|
Upregulation of integrin β4 promotes epithelial-mesenchymal transition and is a novel prognostic marker in pancreatic ductal adenocarcinoma. J Transl Med 2015; 95:308-19. [PMID: 25599535 DOI: 10.1038/labinvest.2014.166] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 11/04/2014] [Accepted: 11/15/2014] [Indexed: 12/27/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a highly aggressive and often lethal malignant tumor. Several studies have shown that epithelial-mesenchymal transition (EMT) is frequently observed in clinical samples of PDA and is related to high metastatic rates and poor outcomes. To identify candidate molecules regulating EMT in PDA, we previously used cDNA microarray analysis and identified integrin β4 (ITGB4) as one of the genes upregulated in high-EMT xenografts derived from PDA patients. The aim of the current study was to clarify the clinicopathological and functional significance of ITGB4 overexpression in PDA. ITGB4 upregulation in high-EMT xenografts was confirmed by immunohistochemistry. Immunohistochemical analyses of 134 surgically resected PDA cases revealed intratumoral heterogeneity with respect to ITGB4 expression and showed that cancer cells undergoing EMT often display strong diffuse ITGB4 expression. High levels of ITGB4 expression were significantly correlated with the hallmarks of EMT (solitary cell infiltration, reduced E-cadherin expression, and increased vimentin expression), with high tumor grade, and with the presence of lymph node metastasis, and showed an independent prognostic effect. Immunocytochemical analyses of PDA cell lines revealed that localization of ITGB4 changed from regions of cell-cell contact to diffuse cytoplasm and cell edges with occasional localization in filopodia during EMT. Knockdown of ITGB4 reduced the migratory and invasive ability of PDA cells. Overexpression of ITGB4 promoted cell scattering and cell motility in combination with downregulation of E-cadherin and upregulation of vimentin expression. In conclusion, we elucidated the prognostic and clinicopathological significance of ITGB4 overexpression in PDA and also the potential role for ITGB4 in the regulation of cancer invasion and EMT.
Collapse
|
75
|
Spina A, De Pasquale V, Cerulo G, Cocchiaro P, Della Morte R, Avallone L, Pavone LM. HGF/c-MET Axis in Tumor Microenvironment and Metastasis Formation. Biomedicines 2015; 3:71-88. [PMID: 28536400 PMCID: PMC5344235 DOI: 10.3390/biomedicines3010071] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/09/2014] [Indexed: 02/07/2023] Open
Abstract
Tumor metastases are responsible for approximately 90% of all cancer-related deaths. Metastasis formation is a multistep process that requires acquisition by tumor cells of a malignant phenotype that allows them to escape from the primary tumor site and invade other organs. Each step of this mechanism involves a deep crosstalk between tumor cells and their microenvironment where the host cells play a key role in influencing metastatic behavior through the release of many secreted factors. Among these signaling molecules, Hepatocyte Growth Factor (HGF) is released by many cell types of the tumor microenvironment to target its receptor c-MET within the cells of the primary tumor. Many studies reveal that HGF/c-MET axis is implicated in various human cancers, and genetic and epigenetic gain of functions of this signaling contributes to cancer development through a variety of mechanisms. In this review, we describe the specific types of cells in the tumor microenvironment that release HGF in order to promote the metastatic outgrowth through the activation of extracellular matrix remodeling, inflammation, migration, angiogenesis, and invasion. We dissect the potential use of new molecules that interfere with the HGF/c-MET axis as therapeutic targets for future clinical trials in cancer disease.
Collapse
Affiliation(s)
- Anna Spina
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| | - Valeria De Pasquale
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| | - Giuliana Cerulo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy.
| | - Pasquale Cocchiaro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy.
| | - Rossella Della Morte
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy.
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy.
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
76
|
Viticchiè G, Muller PAJ. c-Met and Other Cell Surface Molecules: Interaction, Activation and Functional Consequences. Biomedicines 2015; 3:46-70. [PMID: 28536399 PMCID: PMC5344229 DOI: 10.3390/biomedicines3010046] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/08/2015] [Indexed: 12/18/2022] Open
Abstract
The c-Met receptor, also known as the HGF receptor, is one of the most studied tyrosine kinase receptors, yet its biological functions and activation mechanisms are still not fully understood. c-Met has been implicated in embryonic development and organogenesis, in tissue remodelling homeostasis and repair and in cancer metastasis. These functions are indicative of the many cellular processes in which the receptor plays a role, including cell motility, scattering, survival and proliferation. In the context of malignancy, sustained activation of c-Met leads to a signalling cascade involving a multitude of kinases that initiate an invasive and metastatic program. Many proteins can affect the activation of c-Met, including a variety of other cell surface and membrane-spanning molecules or receptors. Some cell surface molecules share structural homology with the c-Met extracellular domain and can activate c-Met via clustering through this domain (e.g., plexins), whereas other receptor tyrosine kinases can enhance c-Met activation and signalling through intracellular signalling cascades (e.g., EGFR). In this review, we provide an overview of c-Met interactions and crosstalk with partner molecules and the functional consequences of these interactions on c-Met activation and downstream signalling, c-Met intracellular localization/recycling and c-Met degradation.
Collapse
Affiliation(s)
- Giuditta Viticchiè
- MRC (Medical Research Council) Toxicology Unit, Lancaster Road, Leicester LE1 9HN, UK.
| | - Patricia A J Muller
- MRC (Medical Research Council) Toxicology Unit, Lancaster Road, Leicester LE1 9HN, UK.
| |
Collapse
|
77
|
Baldanzi G, Graziani A. Physiological Signaling and Structure of the HGF Receptor MET. Biomedicines 2014; 3:1-31. [PMID: 28536396 PMCID: PMC5344233 DOI: 10.3390/biomedicines3010001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/09/2014] [Indexed: 12/13/2022] Open
Abstract
The "hepatocyte growth factor" also known as "scatter factor", is a multifunctional cytokine with the peculiar ability of simultaneously triggering epithelial cell proliferation, movement and survival. The combination of those proprieties results in the induction of an epithelial to mesenchymal transition in target cells, fundamental for embryogenesis but also exploited by tumor cells during metastatization. The hepatocyte growth factor receptor, MET, is a proto-oncogene and a prototypical transmembrane tyrosine kinase receptor. Inhere we discuss the MET molecular structure and the hepatocyte growth factor driven physiological signaling which coordinates epithelial proliferation, motility and morphogenesis.
Collapse
Affiliation(s)
- Gianluca Baldanzi
- Department Translational Medicine, University Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy.
| | - Andrea Graziani
- Department Translational Medicine, University Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy.
- Università Vita-Salute San Raffaele, via Olgettina 58, 20132 Milano, Italy.
| |
Collapse
|
78
|
Molecular architecture and function of the hemidesmosome. Cell Tissue Res 2014; 360:363-78. [PMID: 25487405 PMCID: PMC4544487 DOI: 10.1007/s00441-014-2061-z] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 11/03/2014] [Indexed: 01/07/2023]
Abstract
Hemidesmosomes are multiprotein complexes that facilitate the stable adhesion of basal epithelial cells to the underlying basement membrane. The mechanical stability of hemidesmosomes relies on multiple interactions of a few protein components that form a membrane-embedded tightly-ordered complex. The core of this complex is provided by integrin α6β4 and P1a, an isoform of the cytoskeletal linker protein plectin that is specifically associated with hemidesmosomes. Integrin α6β4 binds to the extracellular matrix protein laminin-332, whereas P1a forms a bridge to the cytoplasmic keratin intermediate filament network. Other important components are BPAG1e, the epithelial isoform of bullous pemphigoid antigen 1, BPAG2, a collagen-type transmembrane protein and CD151. Inherited or acquired diseases in which essential components of the hemidesmosome are missing or structurally altered result in tissue fragility and blistering. Modulation of hemidesmosome function is of crucial importance for a variety of biological processes, such as terminal differentiation of basal keratinocytes and keratinocyte migration during wound healing and carcinoma invasion. Here, we review the molecular characteristics of the proteins that make up the hemidesmosome core structure and summarize the current knowledge about how their assembly and turnover are regulated by transcriptional and post-translational mechanisms.
Collapse
|
79
|
Prat M, Oltolina F, Basilico C. Monoclonal Antibodies against the MET/HGF Receptor and Its Ligand: Multitask Tools with Applications from Basic Research to Therapy. Biomedicines 2014; 2:359-383. [PMID: 28548076 PMCID: PMC5344273 DOI: 10.3390/biomedicines2040359] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibodies can be seen as valuable tools for many aspects of basic as well as applied sciences. In the case of MET/HGFR, they allowed the identification of truncated isoforms of the receptor, as well as the dissection of different epitopes, establishing structure-function relationships. Antibodies directed against MET extracellular domain were found to be full or partial receptor agonists or antagonists. The agonists can mimic the effects of the different isoforms of the natural ligand, but with the advantage of being more stable than the latter. Thus, some agonist antibodies promote all the biological responses triggered by MET activation, including motility, proliferation, morphogenesis, and protection from apoptosis, while others can induce only a migratory response. On the other hand, antagonists can inhibit MET-driven biological functions either by competing with the ligand or by removing the receptor from the cell surface. Since MET/HGFR is often over-expressed and/or aberrantly activated in tumors, monoclonal antibodies can be used as probes for MET detection or as "bullets" to target MET-expressing tumor cells, thus pointing to their use in diagnosis and therapy.
Collapse
Affiliation(s)
- Maria Prat
- Department of Health Sciences, Università del Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy.
| | - Francesca Oltolina
- Department of Health Sciences, Università del Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy.
| | - Cristina Basilico
- Laboratory of Exploratory Research, Candiolo Cancer Institute, Str. Prov. 142, 10060 Candiolo, Italy.
| |
Collapse
|
80
|
Hepatocyte growth factor: A regulator of inflammation and autoimmunity. Autoimmun Rev 2014; 14:293-303. [PMID: 25476732 DOI: 10.1016/j.autrev.2014.11.013] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 11/25/2014] [Indexed: 12/12/2022]
Abstract
Hepatocyte growth factor (HGF) is a pleiotropic cytokine that has been extensively studied over several decades, but was only recently recognized as a key player in mediating protection of many types of inflammatory and autoimmune diseases. HGF was reported to prevent and attenuate disease progression by influencing multiple pathophysiological processes involved in inflammatory and immune response, including cell migration, maturation, cytokine production, antigen presentation, and T cell effector function. In this review, we discuss the actions and mechanisms of HGF in inflammation and immunity and the therapeutic potential of this factor for the treatment of inflammatory and autoimmune diseases.
Collapse
|
81
|
Furlan A, Kherrouche Z, Montagne R, Copin MC, Tulasne D. Thirty Years of Research on Met Receptor to Move a Biomarker from Bench to Bedside. Cancer Res 2014; 74:6737-44. [DOI: 10.1158/0008-5472.can-14-1932] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
82
|
Vay C, Hosch SB, Stoecklein NH, Klein CA, Vallböhmer D, Link BC, Yekebas EF, Izbicki JR, Knoefel WT, Scheunemann P. Integrin expression in esophageal squamous cell carcinoma: loss of the physiological integrin expression pattern correlates with disease progression. PLoS One 2014; 9:e109026. [PMID: 25398092 PMCID: PMC4232252 DOI: 10.1371/journal.pone.0109026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/28/2014] [Indexed: 12/17/2022] Open
Abstract
The integrins are a family of heterodimeric transmembrane signaling receptors that mediate the adhesive properties of epithelial cells affecting cell growth and differentiation. In many epithelial malignancies, altered integrin expression is associated with tumor progression and often correlates with unfavorable prognosis. However, only few studies have investigated the role of integrin expression in esophageal squamous cell carcinoma (ESCC). Using a novel quantifying immunofluorescence-staining assay, we investigated the expression of the integrins α2β1, α3β1, α6β1, and α6β4 in primary ESCC of 36 patients who underwent surgical resection. Magnitude and distribution of expression were analyzed in primary tumor samples and autologous esophageal squamous epithelium. The persistence of the physiologically polarized expression of the subunits α6, β1, and β4 in the tumor tissue was significantly associated with prolonged relapse-free survival (p = 0.028, p = 0.034, p = 0.006). In contrast, patients with reduced focal α6 expression at the tumor invasion front shared a significantly shortened relapse-free survival compared to patients with strong α6 expression at their stromal surfaces, as it was regularly observed in normal esophageal epithelium (p = 0.001). Multivariate regression analysis identified the maintenance of strong α6 immunoreactivity at the invasion front as an independent prognostic factor for increased relapse-free and disease-specific survival (p = 0.003; p = 0.003). Our findings suggest that alterations in both pattern and magnitude of integrin expression may play a major role in the disease progression of ESCC patients. Particularly, the distinct expression of the integrins α6β4 and α6β1 at the invasion front as well as the maintenance of a polarized integrin expression pattern in the tumor tissue may serve as valuable new markers to assess the aggressiveness of ESCC.
Collapse
Affiliation(s)
- Christian Vay
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, Düsseldorf, Germany
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| | - Stefan B. Hosch
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, Düsseldorf, Germany
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Vascular, and Visceral Surgery, Ingolstadt Medical Center, Ingolstadt, Germany
| | - Nikolas H. Stoecklein
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, Düsseldorf, Germany
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph A. Klein
- Division of Oncogenomics, Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Daniel Vallböhmer
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, Düsseldorf, Germany
| | - Björn-Christian Link
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Emre F. Yekebas
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob R. Izbicki
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wolfram T. Knoefel
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, Düsseldorf, Germany
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Scheunemann
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, Düsseldorf, Germany
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Surgery, University Hospital Rostock, Rostock, Germany
| |
Collapse
|
83
|
Wang H, Jin H, Beauvais DM, Rapraeger AC. Cytoplasmic domain interactions of syndecan-1 and syndecan-4 with α6β4 integrin mediate human epidermal growth factor receptor (HER1 and HER2)-dependent motility and survival. J Biol Chem 2014; 289:30318-30332. [PMID: 25202019 PMCID: PMC4215216 DOI: 10.1074/jbc.m114.586438] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/02/2014] [Indexed: 12/14/2022] Open
Abstract
Epithelial cells are highly dependent during wound healing and tumorigenesis on the α6β4 integrin and its association with receptor tyrosine kinases. Previous work showed that phosphorylation of the β4 subunit upon matrix engagement depends on the matrix receptor syndecan (Sdc)-1 engaging the cytoplasmic domain of the β4 integrin and coupling of the integrin to human epidermal growth factor receptor-2 (HER2). In this study, HER2-dependent migration activated by matrix engagement is compared with migration stimulated by EGF. We find that whereas HER2-dependent migration depends on Sdc1, EGF-dependent migration depends on a complex consisting of human epidermal growth factor receptor-1 (HER1, commonly known as EGFR), α6β4, and Sdc4. The two syndecans recognize distinct sites at the extreme C terminus of the β4 integrin cytoplasmic domain. The binding motif in Sdc1 is QEEXYX, composed in part by its syndecan-specific variable (V) region and in part by the second conserved (C2) region that it shares with other syndecans. A cell-penetrating peptide containing this sequence competes for HER2-dependent epithelial migration and carcinoma survival, although it is without effect on the EGFR-stimulated mechanism. β4 mutants bearing mutations specific for Sdc1 and Sdc4 recognition act as dominant negative mutants to block cell spreading or cell migration that depends on HER2 or EGFR, respectively. The interaction of the α6β4 integrin with the syndecans appears critical for it to be utilized as a signaling platform; migration depends on α3β1 integrin binding to laminin 332 (LN332; also known as laminin 5), whereas antibodies that block α6β4 binding are without effect. These findings indicate that specific syndecan family members are likely to have key roles in α6β4 integrin activation by receptor tyrosine kinases.
Collapse
Affiliation(s)
- Haiyao Wang
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Haining Jin
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - DeannaLee M Beauvais
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Alan C Rapraeger
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin 53705; Carbone Cancer Center, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, Wisconsin 53705.
| |
Collapse
|
84
|
Montagne R, Furlan A, Kherrouche Z, Tulasne D. [Thirty years of Met receptor research: from the discovery of an oncogene to the development of targeted therapies]. Med Sci (Paris) 2014; 30:864-73. [PMID: 25311021 DOI: 10.1051/medsci/20143010013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In 1984, the Met receptor and its ligand, the HGF/SF, were discovered thanks to their ability to induce cell transformation and proliferation. Thirty years of research highlighted their crucial role in the development and homeostasis of various structures, including many epithelial organs. This period also allowed unraveling the structural basis of their interaction and their complex signaling network. In parallel, Met was shown to be deregulated and associated with a poor prognosis in many cancers. Met involvement in resistance to current therapies is also being deciphered. Based on these data, pharmaceutical companies developed a variety of Met inhibitors, some of which are evaluated in phase III clinical trials. In this review, we trace the exemplary track record of research on Met receptor, which allowed moving from bench to bedside through the development of therapies targeting its activity. Many questions still remain unanswered such as the involvement of Met in several processes of development, the mechanisms involving Met in resistance to current therapies or the likely emergence of resistances to Met-targeted therapies.
Collapse
Affiliation(s)
- Rémi Montagne
- CNRS UMR 8161, Institut de biologie de Lille, Institut Pasteur de Lille, université de Lille 1 et 2, SIRIC ONCOLille, IFR142, 1, rue du Professeur Calmette, 59021 Lille, France
| | - Alessandro Furlan
- CNRS UMR 8161, Institut de biologie de Lille, Institut Pasteur de Lille, université de Lille 1 et 2, SIRIC ONCOLille, IFR142, 1, rue du Professeur Calmette, 59021 Lille, France
| | - Zoulika Kherrouche
- CNRS UMR 8161, Institut de biologie de Lille, Institut Pasteur de Lille, université de Lille 1 et 2, SIRIC ONCOLille, IFR142, 1, rue du Professeur Calmette, 59021 Lille, France
| | - David Tulasne
- CNRS UMR 8161, Institut de biologie de Lille, Institut Pasteur de Lille, université de Lille 1 et 2, SIRIC ONCOLille, IFR142, 1, rue du Professeur Calmette, 59021 Lille, France
| |
Collapse
|
85
|
Abrantes JLF, Tornatore TF, Pelizzaro-Rocha KJ, de Jesus MB, Cartaxo RT, Milani R, Ferreira-Halder CV. Crosstalk between kinases, phosphatases and miRNAs in cancer. Biochimie 2014; 107 Pt B:167-87. [PMID: 25230087 DOI: 10.1016/j.biochi.2014.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 09/04/2014] [Indexed: 02/07/2023]
Abstract
Reversible phosphorylation of proteins, performed by kinases and phosphatases, is the major post translational protein modification in eukaryotic cells. This intracellular event represents a critical regulatory mechanism of several signaling pathways and can be related to a vast array of diseases, including cancer. Cancer research has produced increasing evidence that kinase and phosphatase activity can be compromised by mutations and also by miRNA silencing, performed by small non-coding and endogenously produced RNA molecules that lead to translational repression. miRNAs are believed to target about one-third of human mRNAs while a single miRNA may target about 200 transcripts simultaneously. Regulation of the phosphorylation balance by miRNAs has been a topic of intense research over the last years, spanning topics going as far as cancer aggressiveness and chemotherapy resistance. By addressing recent studies that have shown miRNA expression patterns as phenotypic signatures of cancers and how miRNA influence cellular processes such as apoptosis, cell cycle control, angiogenesis, inflammation and DNA repair, we discuss how kinases, phosphatases and miRNAs cooperatively act in cancer biology.
Collapse
Affiliation(s)
- Júlia L F Abrantes
- Department of Biochemistry, Institute of Biology, UNICAMP, 13083-970 Campinas, Brazil
| | - Thaís F Tornatore
- Department of Biochemistry, Institute of Biology, UNICAMP, 13083-970 Campinas, Brazil
| | | | - Marcelo B de Jesus
- Department of Biochemistry, Institute of Biology, UNICAMP, 13083-970 Campinas, Brazil
| | - Rodrigo T Cartaxo
- Department of Biochemistry, Institute of Biology, UNICAMP, 13083-970 Campinas, Brazil
| | - Renato Milani
- Department of Biochemistry, Institute of Biology, UNICAMP, 13083-970 Campinas, Brazil
| | | |
Collapse
|
86
|
Liu CH, Hu RH, Huang MJ, Lai IR, Chen CH, Lai HS, Wu YM, Huang MC. C1GALT1 promotes invasive phenotypes of hepatocellular carcinoma cells by modulating integrin β1 glycosylation and activity. PLoS One 2014; 9:e94995. [PMID: 25089569 PMCID: PMC4121071 DOI: 10.1371/journal.pone.0094995] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/21/2014] [Indexed: 02/07/2023] Open
Abstract
Cancer cell invasion and metastasis are the primary causes of treatment failure and death in hepatocellular carcinoma (HCC). We previously reported that core 1 β1,3-galactosyltransferase (C1GALT1) is frequently overexpressed in HCC tumors and its expression is associated with advanced tumor stage, metastasis, and poor survival. However, the underlying mechanisms of C1GALT1 in HCC malignancy remain unclear. In this study, we found that overexpression of C1GALT1 enhanced HCC cell adhesion to extracellular matrix (ECM) proteins, migration, and invasion, whereas RNAi-mediated knockdown of C1GALT1 suppressed these phenotypes. The promoting effect of C1GALT1 on the metastasis of HCC cells was demonstrated in a mouse xenograft model. Mechanistic investigations showed that the C1GALT1-enhanced phenotypic changes in HCC cells were significantly suppressed by anti-integrin β1 blocking antibody. Moreover, C1GALT1 was able to modify O-glycans on integrin β1 and regulate integrin β1 activity as well as its downstream signaling. These results suggest that C1GALT1 could enhance HCC invasiveness through integrin β1 and provide novel insights into the roles of O-glycosylation in HCC metastasis.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/pharmacology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/secondary
- Cell Adhesion/drug effects
- Cell Line, Tumor
- Cell Movement/drug effects
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/metabolism
- Female
- Galactosyltransferases/antagonists & inhibitors
- Galactosyltransferases/genetics
- Galactosyltransferases/metabolism
- Gene Expression Regulation, Neoplastic
- Glycosylation
- Humans
- Integrin beta1/genetics
- Integrin beta1/metabolism
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/pathology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Mice
- Mice, SCID
- Neoplasm Invasiveness
- Polysaccharides/chemistry
- Polysaccharides/metabolism
- Protein Binding
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Chiung-Hui Liu
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Rey-Heng Hu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Miao-Juei Huang
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - I-Rue Lai
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Hua Chen
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hong-Shiee Lai
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yao-Ming Wu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail: (M-CH); (Y-MW)
| | - Min-Chuan Huang
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail: (M-CH); (Y-MW)
| |
Collapse
|
87
|
Nanoparticle-formulated siRNA targeting integrins inhibits hepatocellular carcinoma progression in mice. Nat Commun 2014; 5:3869. [PMID: 24844798 PMCID: PMC4107318 DOI: 10.1038/ncomms4869] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 04/11/2014] [Indexed: 02/07/2023] Open
Abstract
Integrins play an important role during development, regulating cell differentiation, proliferation and survival. Here we show that knockdown of integrin subunits slows down the progression of hepatocellular carcinoma (HCC). Using nanoparticulate delivery of short interfering RNAs targeting β1 and αv integrin subunits we downregulate all integrin receptors in hepatocytes. Short-term integrin knockdown (two weeks) does not cause apparent structural or functional perturbations of normal liver tissue. Alterations in liver morphology accumulate upon sustained integrin downregulation (seven weeks). The integrin knockdown leads to significant retardation of HCC progression, reducing proliferation and increasing tumour cell death. This tumour retardation is accompanied by reduced activation of MET oncogene as well as expression of its mature form on the cell surface. Our data suggest that transformed proliferating cells from HCC are more sensitive to knockdown of integrins than normal quiescent hepatocytes, highlighting the potential of siRNA-mediated inhibition of integrins as an anti-cancer therapeutic approach.
Collapse
|
88
|
Gentile A, Lazzari L, Benvenuti S, Trusolino L, Comoglio PM. The ROR1 pseudokinase diversifies signaling outputs in MET-addicted cancer cells. Int J Cancer 2014; 135:2305-16. [DOI: 10.1002/ijc.28879] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 02/21/2014] [Accepted: 03/20/2014] [Indexed: 01/07/2023]
Affiliation(s)
- Alessandra Gentile
- ECMO, Experimental Clinical Molecular Oncology, Candiolo Cancer Institute-FPO, IRCCS; Candiolo (Torino) Italy
| | - Luca Lazzari
- ECMO, Experimental Clinical Molecular Oncology, Candiolo Cancer Institute-FPO, IRCCS; Candiolo (Torino) Italy
| | - Silvia Benvenuti
- ECMO, Experimental Clinical Molecular Oncology, Candiolo Cancer Institute-FPO, IRCCS; Candiolo (Torino) Italy
| | - Livio Trusolino
- ECMO, Experimental Clinical Molecular Oncology, Candiolo Cancer Institute-FPO, IRCCS; Candiolo (Torino) Italy
- Department of Oncology; University of Torino School of Medicine; Torino Italy
| | - Paolo Maria Comoglio
- ECMO, Experimental Clinical Molecular Oncology, Candiolo Cancer Institute-FPO, IRCCS; Candiolo (Torino) Italy
- Department of Oncology; University of Torino School of Medicine; Torino Italy
| |
Collapse
|
89
|
Martin LP, Sill M, Shahin MS, Powell M, DiSilvestro P, Landrum LM, Gaillard SL, Goodheart MJ, Hoffman J, Schilder RJ. A phase II evaluation of AMG 102 (rilotumumab) in the treatment of persistent or recurrent epithelial ovarian, fallopian tube or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol 2014; 132:526-30. [PMID: 24361733 PMCID: PMC4469031 DOI: 10.1016/j.ygyno.2013.12.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/05/2013] [Accepted: 12/13/2013] [Indexed: 01/09/2023]
Abstract
OBJECTIVE This open-label, multi-institutional phase II trial evaluated activity and safety of rilotumumab (AMG 102), a monoclonal antibody that targets HGF (hepatocyte growth factor), the ligand for the MET receptor, in women with recurrent or persistent epithelial ovarian, fallopian tube or primary peritoneal cancer. PATIENTS AND METHODS Women were eligible for treatment with rilotumumab if they had measurable disease, a performance status of 0, 1 or 2, previously received platinum-based therapy with a progression-free interval of <12 months or a second recurrence, and adequate bone marrow and organ function. Patients received rilotumumab 20mg/kg IV every 14 days until evidence of unacceptable toxicity or disease progression. The study utilized co-dual primary endpoints of tumor response and six-month PFS to assess the efficacy of rilotumumab. Secondary endpoints included the frequency and severity of adverse events and the duration of progression-free and overall survival. RESULTS Thirty-one women enrolled and received rilotumumab. All were eligible for analysis. One patient achieved a complete response (3.2%; 90% CI 0.2-14%), and two women had 6-month PFS (6.5%; 90% CI 1.1-19%). Most adverse events were grade 1 or 2, with no grade 4 adverse events. Grade 3 adverse events were gastrointestinal (4), metabolic (3) anemia (3), a thromboembolic event (1), ventricular tachycardia (1), hypotension during infusion (1) and fatigue (1). The study was stopped after the first stage of accrual. CONCLUSION Rilotumumab was well-tolerated, but had limited activity. The level of activity does not warrant further evaluation of rilotumumab as a single agent in patients with ovarian cancer.
Collapse
Affiliation(s)
- Lainie P Martin
- Dept. of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | - Michael Sill
- Gynecologic Oncology Group Statistical & Data Center, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Mark S Shahin
- Dept. of Clinical Gynecologic Oncology, Hanjani Institute for Gynecologic Oncology, Abington Memorial Hospital, Abington, PA 19001, USA
| | - Matthew Powell
- Dept. of OB/GYN, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Paul DiSilvestro
- Program in Women's Oncology, Women & Infants Hospital/Alpert School of Medicine, Providence, RI 02905, USA
| | - Lisa M Landrum
- Dept. of OB/GYN, Oklahoma University Health Science Center, Oklahoma City, OK 73104, USA
| | - Stephanie L Gaillard
- Dept. of Gynecologic Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Michael J Goodheart
- Dept. of Gynecologic Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - James Hoffman
- Dept. of Gynecologic Oncology, The Hospital of Central Connecticut, New Britain, CT 06050, USA
| | - Russell J Schilder
- Dept. of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
90
|
Giordano S, Columbano A. Met as a therapeutic target in HCC: facts and hopes. J Hepatol 2014; 60:442-52. [PMID: 24045150 DOI: 10.1016/j.jhep.2013.09.009] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/13/2013] [Accepted: 09/03/2013] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide, and its burden is expected to increase further in the next years. In spite of the advances of classical therapies, such as surgery, transplantation, use of radiofrequency and transarterial embolization, the prognosis of this neoplasm has not considerably improved over the past few years. The advent of targeted therapies and the approval of the systemic treatment of advanced HCC with the kinase inhibitor sorafenib have provided some hope for the future. Even if the molecular mechanisms responsible for the onset and progression of HCC are still largely unknown, new therapeutic targets have recently come to the spotlight. One of these targets is the tyrosine kinase receptor for the Hepatocyte Growth Factor, encoded by the MET gene, known to promote tumor growth and metastasis in many human organs. In this review we will summarize the contrasting results obtained in vitro (in HCC cell lines) and in animal experimental models and we will also try to analyze the reasons for the opposite findings, suggesting that the HGF/MET axis can have either a promoting or a suppressive role in the development of HCC. We will also reconsider the evidence of activation of this pathway in human HCCs and discuss the results of the clinical trials performed with MET inhibitors. The final purpose is to better clarify which can be the role of MET as a therapeutic target in HCC.
Collapse
Affiliation(s)
- Silvia Giordano
- Department of Oncology, University of Torino, Institute for Cancer Research and Treatment (IRCC), 10060 Candiolo (Torino), Italy.
| | - Amedeo Columbano
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
91
|
Xu Y, Fisher GJ. Role of met axis in head and neck cancer. Cancers (Basel) 2013; 5:1601-18. [PMID: 24287743 PMCID: PMC3875956 DOI: 10.3390/cancers5041601] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/12/2013] [Accepted: 11/14/2013] [Indexed: 01/30/2023] Open
Abstract
Head and neck cancer is the sixth most common type of cancer worldwide. Despite advances in aggressive multidisciplinary treatments, the 5-year survival rate for this dreadful disease is only 50%, mostly due to high rate of recurrence and early involvement of regional lymph nodes and subsequent metastasis. Understanding the molecular mechanisms responsible for invasion and metastasis is one of the most pressing goals in the field of head and neck cancer. Met, also known as hepatocyte growth factor receptor (HGFR), is a member of the receptor protein tyrosine kinase (RPTK) family. There is compelling evidence that Met axis is dysregulated and plays important roles in tumorigenesis, progression, metastasis, angiogenesis, and drug resistance in head and neck cancer. We describe in this review current understanding of Met axis in head and neck cancer biology and development of therapeutic inhibitors targeting Met axis.
Collapse
Affiliation(s)
- Yiru Xu
- Authors to whom correspondence should be addressed; E-Mails: (Y.X.); (G.J.F.); Tel.: +1-734-763-1469 (G.J.F.); Fax: +1-734-647-0076 (G.J.F.)
| | - Gary J. Fisher
- Authors to whom correspondence should be addressed; E-Mails: (Y.X.); (G.J.F.); Tel.: +1-734-763-1469 (G.J.F.); Fax: +1-734-647-0076 (G.J.F.)
| |
Collapse
|
92
|
Soung YH, Korneeva N, Kim TH, Chung J. The role of c-Src in integrin (α6β4) dependent translational control. BMC Cell Biol 2013; 14:49. [PMID: 24180592 PMCID: PMC4228388 DOI: 10.1186/1471-2121-14-49] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 10/25/2013] [Indexed: 11/10/2022] Open
Abstract
Background Integrin α6β4 contributes to cancer progression by stimulating transcription as well as translation of cancer related genes. Our previous study demonstrated that α6β4 stimulates translation initiation of survival factors such as VEGF by activating mTOR pathway. However, the immediate early signaling events that link α6β4 to mTOR activation needs to be defined. Results In the current studies, we demonstrated that c-Src is an immediate early signaling molecule that acts upstream of α6β4 dependent mTOR activation and subsequent translation of VEGF in MDA-MB-435/β4 and MDA-MB-231 cancer cells. m7GTP-Sepharose–binding assay revealed that Src activity is required to form eIF4F complex which is necessary for Cap-dependent translation in α6β4 expressing human cancer cells. Conclusions Overall, our studies suggest that integrin β4 and c-Src activation is important early signaling events to lead mTOR activation and cap-dependent translation of VEGF.
Collapse
Affiliation(s)
| | | | | | - Jun Chung
- Department of Physiology and Stephenson Cancer Center, The University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA.
| |
Collapse
|
93
|
Yamamoto N, Murakami H, Hayashi H, Fujisaka Y, Hirashima T, Takeda K, Satouchi M, Miyoshi K, Akinaga S, Takahashi T, Nakagawa K. CYP2C19 genotype-based phase I studies of a c-Met inhibitor tivantinib in combination with erlotinib, in advanced/metastatic non-small cell lung cancer. Br J Cancer 2013; 109:2803-9. [PMID: 24169346 PMCID: PMC3844902 DOI: 10.1038/bjc.2013.588] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 08/23/2013] [Accepted: 09/04/2013] [Indexed: 12/15/2022] Open
Abstract
Background: A previous clinical study in non-small cell lung cancer (NSCLC) patients in Western countries suggested the potential for combination of a first-in-class non-ATP-competitive c-Met inhibitor tivantinib with an epidermal growth factor receptor-tyrosine kinase inhibitor erlotinib. Polymorphisms of CYP2C19, the key metabolic enzyme for tivantinib, should be addressed to translate the previous Western study to Asian population, because higher incidence of poor metabolisers (PMs) is reported in Asian population. Methods: Japanese patients with advanced/metastatic NSCLC received tivantinib in combination with erlotinib to evaluate safety and pharmacokinetics. Doses of tivantinib were escalated separately for extensive metabolisers (EMs) and PMs. Results: Tivantinib, when combined with erlotinib, was well tolerated up to 360 mg BID for EMs and 240 mg BID for PMs, respectively. Among 25 patients (16 EMs and 9 PMs), the adverse events (AEs) related to tivantinib and/or erlotinib (>20%, any grade) were rash, diarrhoea, dry skin and nausea. Grade ⩾3 AEs were leukopenia, anaemia and neutropenia. No dose-limiting toxicity was observed. Pharmacokinetics profile of tivantinib was not clearly different between the combination and monotherapy. Three partial response and three long-term stable disease (⩾24 weeks) were reported. Conclusion: Two doses of tivantinib in combination with erlotinib were recommended based on CYP2C19 genotype: 360 mg BID for EMs and 240 mg BID for PMs.
Collapse
Affiliation(s)
- N Yamamoto
- Division of Thoracic Oncology, Shizuoka Cancer Center, 1007, Shimonagakubo, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Kinoshita T, Nohata N, Hanazawa T, Kikkawa N, Yamamoto N, Yoshino H, Itesako T, Enokida H, Nakagawa M, Okamoto Y, Seki N. Tumour-suppressive microRNA-29s inhibit cancer cell migration and invasion by targeting laminin-integrin signalling in head and neck squamous cell carcinoma. Br J Cancer 2013; 109:2636-45. [PMID: 24091622 PMCID: PMC3833206 DOI: 10.1038/bjc.2013.607] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/27/2013] [Accepted: 09/12/2013] [Indexed: 01/22/2023] Open
Abstract
Background: Our recent studies of microRNA (miRNA) expression signatures demonstrated that microRNA-29s (miR-29s; miR-29a/b/c) were significantly downregulated in head and neck squamous cell carcinoma (HNSCC) and were putative tumour-suppressive miRNAs in human cancers. Our aim in this study was to investigate the functional significance of miR-29s in cancer cells and to identify novel miR-29s-mediated cancer pathways and responsible genes in HNSCC oncogenesis and metastasis. Methods: Gain-of-function studies using mature miR-29s were performed to investigate cell proliferation, migration and invasion in two HNSCC cell lines (SAS and FaDu). To identify miR-29s-mediated molecular pathways and targets, we utilised gene expression analysis and in silico database analysis. Loss-of-function assays were performed to investigate the functional significance of miR-29s target genes. Results: Restoration of miR-29s in SAS and FaDu cell lines revealed significant inhibition of cancer cell migration and invasion. Gene expression data and in silico analysis demonstrated that miR-29s modulated the focal adhesion pathway. Moreover, laminin γ2 (LAMC2) and α6 integrin (ITGA6) genes were candidate targets of the regulation of miR-29s. Luciferase reporter assays showed that miR-29s directly regulated LAMC2 and ITGA6. Silencing of LAMC2 and ITGA6 genes significantly inhibited cell migration and invasion in cancer cells. Conclusion: Downregulation of miR-29s was a frequent event in HNSCC. The miR-29s acted as tumour suppressors and directly targeted laminin–integrin signalling. Recognition of tumour-suppressive miRNA-mediated cancer pathways provides new insights into the potential mechanisms of HNSCC oncogenesis and metastasis and suggests novel therapeutic strategies for the disease.
Collapse
Affiliation(s)
- T Kinoshita
- 1] Department of Functional Genomics, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan [2] Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Thuma F, Ngora H, Zöller M. The metastasis-associated molecule C4.4A promotes tissue invasion and anchorage independence by associating with the alpha6beta4 integrin. Mol Oncol 2013; 7:917-28. [PMID: 23727360 PMCID: PMC5528461 DOI: 10.1016/j.molonc.2013.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 05/01/2013] [Accepted: 05/02/2013] [Indexed: 01/13/2023] Open
Abstract
C4.4A is a metastasis-associated molecule that functions appear to rely on associated alph6beta4 integrin. To corroborate the impact of the C4.4A-alpha6beta4 integrin association on metastasis formation, C4.4A was knocked-down in a highly metastatic rat pancreatic adenocarcinoma (ASML, ASML-C4.4Akd). Metastasis formation by ASML-C4.4Akd cells after intrafootpad application was strongly retarded in draining nodes and lung colonization was rare. Furthermore, cisplatin treatment significantly prolonged the survival time only of ASML-C4.4Akd-bearing rats. ASML-C4.4Akd cells display reduced migratory activity and impaired matrix protein degradation due to inefficient MMP14 activation; loss of drug-resistance is due to mitigated PI3K/Akt pathway activation. These losses of function rely on the laminin receptor C4.4A recruiting activated alpha6beta4 integrin into rafts, where C4.4A cooperates with alpha6beta4 and via alpha6beta4 with MMP14. Within this raft-located complex, MMP14 provokes focalized matrix degradation and mostly alpha6beta4 integrin promotes BAD phosphorylation and upregulated Bcl2 and BclXl expression. Thus, metastasis-promoting activities of C4.4A are not genuine characteristics of C4.4A. Instead, the raft-located laminin receptor C4.4A recruits alpha6beta4 integrin and supports via the alpha6beta4 integrin MMP14 activation. Thereby C4.4A acts as a linker to facilitate several steps in the metastatic cascade. Taking the restricted C4.4A expression in non-transformed tissue, this knowledge should pave the way toward the use of C4.4A as a therapeutic target.
Collapse
Affiliation(s)
- Florian Thuma
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg D-69120, Germany
| | - Honoré Ngora
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg D-69120, Germany
| | - Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg D-69120, Germany
- German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
96
|
Corso S, Giordano S. Cell-autonomous and non-cell-autonomous mechanisms of HGF/MET-driven resistance to targeted therapies: from basic research to a clinical perspective. Cancer Discov 2013; 3:978-92. [PMID: 23901039 DOI: 10.1158/2159-8290.cd-13-0040] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
UNLABELLED Targeted therapies have opened new perspectives in clinical oncology. However, clinicians have observed a lack of response in a relevant percentage of patients and frequent relapse in patients who initially respond. Therefore, a compelling challenge is to identify mechanisms underlying resistance and strategies to circumvent these hurdles. A growing body of evidence indicates that MET, the tyrosine kinase receptor for hepatocyte growth factor (HGF), is frequently implicated in resistance to targeted therapies. In this review, we highlight cell-autonomous and non-cell-autonomous mechanisms through which MET drives resistance, and we discuss some unsolved issues related to the selection of patients who could benefit from combined therapies. SIGNIFICANCE Resistance is, at present, the major limitation to the efficacy of targeted therapies. Inappropriate MET activation is very frequently implicated in the onset of primary and secondary resistance to these therapies. Deciphering the role of the HGF/MET axis in resistance to different drugs could guide the design of new clinical trials based on combinatorial therapies, and it might help to overcome, or possibly prevent, the onset of resistance.
Collapse
Affiliation(s)
- Simona Corso
- Department of Oncology, Institute for Cancer Research at Candiolo, University of Torino, Candiolo, Torino, Italy
| | | |
Collapse
|
97
|
Abstract
Integrins are transmembrane receptors that mediate cell adhesion to neighboring cells and to the extracellular matrix. Here, the various modes in which integrin-mediated adhesion regulates intracellular signaling pathways impinging on cell survival, proliferation, and differentiation are considered. Subsequently, evidence that integrins also control crucial signaling cascades in cancer cells is discussed. Lastly, the important role of integrin signaling in tumor cells as well as in stromal cells that support cancer growth, metastasis, and therapy resistance indicates that integrin signaling may be an attractive target for (combined) cancer therapy strategies. Current approaches to target integrins in this context are reviewed.
Collapse
|
98
|
Iwata H, Kitajima Y. Bullous pemphigoid: role of complement and mechanisms for blister formation within the lamina lucida. Exp Dermatol 2013; 22:381-5. [PMID: 23651418 DOI: 10.1111/exd.12146] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2013] [Indexed: 01/15/2023]
Abstract
Bullous pemphigoid (BP), an autoimmune subepidermal blistering skin disease, demonstrates tense blisters with or without widespread erythema, blistering along the lamina lucida, immunoglobulin G and/or complement deposits at the basement membrane zone, and the presence of circulating autoantibodies against hemidesmosomal molecules. These autoantibodies usually react against 180-kDa and/or 230-kDa proteins, designated as BP180 and BP230, respectively. The precise blistering mechanisms after autoantibodies bind to antigens are not fully understood. Immune complexes are thought to initially activate the complement cascade, which may induce activation of proteases and/or cytokines and cause dermal-epidermal separation. However, why does separation run specifically within the lamina lucida in a space as narrow as 500 nm wide? This review mainly focuses on the possible mechanisms of BP-specific blistering and how separation occurs along the lamina lucida, based on existing evidence.
Collapse
Affiliation(s)
- Hiroaki Iwata
- Department of Dermatology, University of Lübeck, Lübeck, Germany.
| | | |
Collapse
|
99
|
Ding YB, Deng B, Huang YS, Xiao WM, Wu J, Zhang YQ, Wang YZ, Wu DC, Lu GT, Wu KY. A high level of integrin α6 expression in human intrahepatic cholangiocarcinoma cells is associated with a migratory and invasive phenotype. Dig Dis Sci 2013; 58:1627-1635. [PMID: 23306848 DOI: 10.1007/s10620-012-2524-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 12/03/2012] [Indexed: 02/05/2023]
Abstract
BACKGROUND The integrin α6 subunit is part of the integrin α6β1 and α6β4 complexes, which are known to mediate the invasion of carcinoma cells. However, the precise role of integrin α6 in intrahepatic cholangiocarcinoma (ICC) has not yet been addressed. METHODS Twenty cases of ICCs and matched nontumor samples were used to analyze integrin α6 expression by immunohistochemistry. After the expression of integrin α6 was determined by RT-PCR and Western blot in ICC cells, we regulated the expression of integrin α6 in ICC cells with specific vshRNA-integrin α6, and assessed the role of integrin α6 in the proliferation and metastasis/invasion of ICC cells. Finally, the involved mechanisms and clinical significance were further investigated. RESULTS The expression of integrin α6 in ICC tissues was much higher than that in nontumor samples, and the high level of integrin α6 was detected in ICC cells compared with normal liver cells and HepG2 cells. After the down-regulation of integrin α6 in HCCC-9810 cells, we showed that the ability of ICC cells to metastasize and invade was much decreased in vitro, and cell proliferation was inhibited significantly. Further study indicated high expression of integrin α6 enhanced the activation of ERK1/2 and AKT signals in ICC cells and the inhibition of ERK1/2 down-regulated ICC cell proliferation, while the inhibition of AKT markedly impaired ICC cell metastasis and invasion. Integrin α6 overexpression was significantly correlated with larger tumors, multiple nodular, microvascular/bile duct invasion, and lymphatic metastasis (p < 0.05). The postoperative 5-year overall survival (OS) rate in patients with integrin α6(low) was higher than that of the integrin α6(high) group. CONCLUSIONS Overexpression of integrin α6 is associated with a migratory and invasive phenotype of ICC, and integrin α6 may be used as molecular target for therapy of ICC.
Collapse
Affiliation(s)
- Yan-bing Ding
- Department of Gastroenterology, Yangzhou No. 1 People's Hospital, The Second Clinical School of Yangzhou University, Yangzhou, 225000, People's Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Hasenauer S, Malinger D, Koschut D, Pace G, Matzke A, von Au A, Orian-Rousseau V. Internalization of Met requires the co-receptor CD44v6 and its link to ERM proteins. PLoS One 2013; 8:e62357. [PMID: 23626807 PMCID: PMC3633891 DOI: 10.1371/journal.pone.0062357] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 03/20/2013] [Indexed: 12/14/2022] Open
Abstract
Receptor Tyrosine Kinases (RTKs) are involved in many cellular processes and play a major role in the control of cell fate. For these reasons, RTK activation is maintained under tight control. Met is an essential RTK that induces proliferation, differentiation, migration, survival and branching morphogenesis. Deregulation of Met by overexpression, amplification or lack of effective degradation leads to cancer and metastasis. We have shown that Met relies on CD44v6 for its activation and for signaling in several cancer cell lines and also in primary cells. In this paper, we show that internalization of Met is dependent on CD44v6 and the binding of Ezrin to the CD44v6 cytoplasmic domain. Both CD44v6 and Met are co-internalized upon Hepatocyte Growth Factor induction suggesting that Met-induced signaling from the endosomes relies on its collaboration with CD44v6 and the link to the cytoskeleton provided by ERM proteins.
Collapse
Affiliation(s)
- Susanne Hasenauer
- Karlsruhe Institute of Toxicology, Institute for Toxicology and Genetics, Karlsruhe, Germany
| | - Dieter Malinger
- Karlsruhe Institute of Toxicology, Institute for Toxicology and Genetics, Karlsruhe, Germany
| | - David Koschut
- Karlsruhe Institute of Toxicology, Institute for Toxicology and Genetics, Karlsruhe, Germany
| | - Giuseppina Pace
- Karlsruhe Institute of Toxicology, Institute for Toxicology and Genetics, Karlsruhe, Germany
| | - Alexandra Matzke
- Karlsruhe Institute of Toxicology, Institute for Toxicology and Genetics, Karlsruhe, Germany
| | - Anja von Au
- Karlsruhe Institute of Toxicology, Institute for Toxicology and Genetics, Karlsruhe, Germany
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Toxicology, Institute for Toxicology and Genetics, Karlsruhe, Germany
- * E-mail:
| |
Collapse
|