1. Trang chủ
  2. » Giáo án - Bài giảng

gene of the month hgf

5 1 0

Đang tải... (xem toàn văn)

THÔNG TIN TÀI LIỆU

Thông tin cơ bản

Định dạng
Số trang 5
Dung lượng 412,61 KB

Nội dung

Gene of the month Gene of the month: HGF Ana Belen Fajardo-Puerta,1 Mireia Mato Prado,2 Adam E Frampton,1,2 Long R Jiao1 HPB Surgical Unit, Department of Surgery & Cancer, Imperial College, London, UK Division of Cancer, Department of Surgery & Cancer, Imperial College, London, UK Correspondence to Prof Long R Jiao, HPB Surgical Unit, Department of Surgery & Cancer, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London W12 0HS, UK; l.jiao@imperial.ac.uk Accepted 21 March 2016 Published Online First 12 April 2016 ABSTRACT Hepatocyte growth factor (HGF) is a multifunctional cytokine with important roles in cell proliferation, survival, motility and morphogenesis Secreted by cells of mesenchymal origin, HGF is the specific ligand for the tyrosine-kinase receptor c-MET (cellular mesenchymal-epithelial transition), also called MET, which is expressed in different types of epithelial, endothelial and haematopoietic progenitor cells The HGF/MET axis is involved in several biological processes, such as embryogenesis, organogenesis, adult tissue regeneration (including wound healing and liver regeneration) and carcinogenesis, for both solid and haematological malignancies.1 HGF and its particular interaction with the MET receptor have been extensively investigated in the last decades and remain the focus of numerous clinical trials.3–8 This short review focuses on HGF structure and function, as well as its roles in liver regeneration and different types of tumours HGF STRUCTURE To cite: Fajardo-Puerta AB, Mato Prado M, Frampton AE, et al J Clin Pathol 2016;69:575–579 HGF is mapped on the long arm of chromosome at q21.1 and it is formed by 18 exons, interrupted by 17 introns, spanning 71 433 bases of genomic DNA It encodes the inactive pre-pro-HGF, a single chain of 728 amino acids (83 134 Da), which includes a signal sequence (1–31), a heavy α chain (32–494; 69 kDa) and a light β chain (495–728; 34 kDa) The first exon contains the signal peptide and a 50 -untranslated region The following 10, 12 and remaining exons encode the α chain, with four kringle structures, the short spacer region between the α and β chains, and the β chain, respectively.9–11 The HGF primary structure was determined in 1989, but multiple transcript variants encoding different isoforms have been identified by alternative splicing of the gene since then.12 The inactive pre-pro-HGF becomes active after a two-cleavage process First, the signal peptide of the pre-pro-HGF is degraded, generating the pro-HGF, which is also cleaved between Arg494 and Val495 Several serum or cell-membrane proteases have been described to be involved, such as HGF activator (HGF-A), urokinase-type plasminogen activator, plasma kallikrein, coagulation factors XII and XI, matriptase and hepsin.13 Among them, HGF-A is the main protease responsible for the activation of pro-HGF in serum The activation process of HGF plays an important role in the regulation of tissue regeneration and susceptibility to pathological conditions Although HGF-A knockout mice showed normal development, they have impaired restitution of epithelia after mucosal injury.14 Similarly, fibroblasts from patients with idiopathic pulmonary fibrosis have been shown to have lower capacity to activate HGF compared with control fibroblasts.15 The final active heterodimeric molecule is produced by a disulfide bond between the α and β chains Although this multifunctional protein belongs to the plasminogen subfamily of S1 peptidases, no proteolytic activity has been detected, probably due to the substitution of two out of three amino acids required in the catalytic triad.16 It has been suggested that HGF could be evolutionally derived from those encoding proteases implicated in the coagulation cascade and fibrinolysis Indeed, its organisation is very similar, and so are its products The HGF α chain has 38% homology with plasmin, and the β chain is structurally similar to the catalytic domains of serine proteases, but with amino acid substitutions in the active site.1 In contrast to other growth factors and their receptors, HGF binds exclusively to the product of the c-MET proto-oncogene, also mapped on chromosome 7.17–19 The MET receptor is a 190 kDa protein, comprising a ligand-binding extracellular domain, a transmembrane region and a cytoplasmic domain with tyrosine kinase activity Although the HGF α chain has a higher affinity for MET, it is the β chain which activates the receptor.20 21 Upon MET dimerisation, kinase activation results in autophosphorylation of tyrosines Y1349 and Y1356, and recruitment of several substrates, including growth factor receptor-bound protein 2, Shc, p85 subunit of phosphatidylinositol 30 kinase, phospholipase C γ, signal transducer and activator of transcription and Grb2-associated binding protein The downstream signalling pathways generate diverse cellular responses, such as proliferation, survival, motility, invasion and stimulation of angiogenesis (figure 1).22–34 Several growth factors, cytokines and prostaglandins upregulate HGF gene expression: basic fibroblast growth factor, oncostatin M, hypoxiainducible factor α and nuclear factor κ-B Its main inhibitor is tumour growth factor β.35 HGF FUNCTION AND ITS RECEPTOR In 1984, HGF was purified for the first time in rat platelets, and years later in humans It was described as a potent mitogenic factor for mature rat hepatocytes in vitro.36–39 The HGF/MET pathway has diverse biological and physiological roles in organogenesis, morphogenesis, tissue regeneration and carcinogenesis that have been discovered using conditional knockout of MET in mice.8 In 1991, a fibroblast-derived factor for epithelial cells, the scatter factor, involved in increasing cell migration during embryogenesis and tumour progression, together with the human lung fibroblast-derived mitogen, were found to be identical proteins Both are encoded by the same chromosome bands as HGF and are ligands for the MET receptor.40–44 Soon after, a tumour cytotoxic Fajardo-Puerta AB, et al J Clin Pathol 2016;69:575–579 doi:10.1136/jclinpath-2015-203575 575 Gene of the month Figure The hepatocyte growth factor/mesenchymal-epithelial transition (HGF/MET) axis Once HGF is activated by a two-cleavage process in the extracellular matrix, it binds to the MET receptor on epithelial cells, promoting its dimerisation and autophosphorylation of tyrosine residues The recruitment of adaptor proteins generates different downstream signalling pathways which evoke diverse cellular responses as shown C-Cbl, casitas b-lineage lymphoma; Erk1-2, extracellular signal-regulated kinases; Gab1, Grb2-associated binding protein 1; Grb2, growth factor receptor bound protein 2; Gsk3β, glycogen synthetase kinase 3β; IKK, inhibitor of nuclear factor κ-B kinase; MEK, mitogen-activated protein kinase/ERK kinase; mTor, mammalian target of rapamycin; NF-kβ, nuclear factor κ-B; PI3K, phosphatidylinositol 30 kinase; Plcγ, phospholipase C-γ; Stat3, signal transducer and activator of transcription (Adapted from Boissinot et al35) factor derived from fibroblasts, which induces cell death in several cancer types, was also found to be an identical molecule to HGF.45 These results had important consequences for further studies on the involvement of HGF as a modulator of cellular growth and motility during embryogenesis, tissue regeneration and carcinogenesis Interestingly, targeted disruption of HGF or MET results in embryonically lethal knockouts with impaired development of the liver and placenta.46 47 Overexpression, mutations and amplification of the receptor, and/or changes in its kinase activity have been implicated in different types of cancer.48 It has been suggested that because of the proximity between HGF and MET on chromosome 7, a polysomy of this chromosome could lead to malignancy secondary to overproduction of both molecules.49 The one-to-one ligand-receptor relationship makes the HGF/MET axis an attractive target for drug development, either by activation or inhibition of this pathway Three pharmacological approaches are currently being developed as inhibitors of MET signalling with promising results: anti-HGF antibodies, anti-MET antibodies and MET kinase inhibitors.50 HGF AND LIVER REGENERATION The liver has the ability to regenerate to almost its optimal volume after liver resection.51 HGF is one of two complete mitogens which induce hepatocyte DNA synthesis and mitosis along with epidermal growth factor receptor (EGFR) Activation of the HGF/MET axis generates a cascade of intracellular signalling for the G1-S progression of hepatocytes Available evidence has shown that immediately after different liver injuries, such as partial hepatectomy, ischaemia or hepatitis, there is an intense 576 remodelling of the extracellular matrix, with increased activity of proteinases and intense intracellular signalling.52 The levels of active HGF rise rapidly in the liver secondary to its production by Kupffer, stellate and sinusoidal endothelial cells, and subsequently activation by urokinases.53–55 Since HGF also increases in plasma, an endocrine mechanism has been suggested In addition, HGF transcript levels and HGF activity have been found to be markedly higher in other intact organs like the lung, kidney or spleen after injuries of the liver.56 Overall, this results in balanced liver growth and regeneration HGF AND HEPATOCELLULAR CARCINOMA Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related mortality worldwide with a multifactorial aetiology and extensive molecular and phenotypical heterogeneity.57 Several signalling pathways have been described in which the HGF/MET axis may play a crucial role.58–60 Patients with HCC have significantly higher serum levels of HGF compared with healthy controls52 61 and overexpression of HGF and c-MET has been detected in 33% and 20%–48% of HCC tissues, respectively.62–66 Upregulation of MET has also been associated with tumour migration, vascular invasion, neoangiogenesis and, therefore, poor patient outcomes.67 In contrast, other studies have shown contradictory results with regard to their impact on survival in patients with HCC.68–73 Nonetheless, clinical trials using pharmacological inhibitors of this axis for both HCC and other solid malignancies are currently being performed These targeted therapies could be a promising second line option to treat patients with advanced HCC.60 74 75 A recent study has demonstrated that the Fajardo-Puerta AB, et al J Clin Pathol 2016;69:575–579 doi:10.1136/jclinpath-2015-203575 Gene of the month mixed-lineage leukaemia protein (MLL), in association with HGF/MET, promotes cell invasion and metastasis in HCC Theoretically, the inhibition of this interaction could potentially reduce the incidence of distant metastasis, although it may not affect the tumour load or its proliferative capacity.76 HGF IN OESOPHAGOGASTRIC CANCER Oesophagogastric cancer (OGC) is the fifth most common malignancy worldwide and the fourth and fifth most common type of cancer death in men and women in UK, respectively A clear geographical difference has been observed in overall survival with 70% surviving years in Japan, compared with 25% in Europe, suggesting that the implementation of screening tools might allow earlier detection and treatment of this cancer OGC has a diverse molecular landscape and exhibits alterations in various different oncogenes and kinase pathways Activation of the HGF/MET axis promoting tumourigenesis and metastasis in gastric cancer is mainly secondary to MET overexpression and/ or amplification, which was observed in 75%–90% of cases and in 1.5%–20% of the patients, respectively.77–79 However, mutations of HGF or MET are extremely rare in OGC.80 81 Different drugs targeting the axis include tyrosine kinase inhibitors (crizotinib, a dual c-MET and ALK (anaplastic lymphoma kinase) inhibitor), and monoclonal antibodies that neutralise HGF (rilotumumab) or MET (onartuzumab) are currently being trialled.82–86 HGF IN COLORECTAL CANCER Colorectal cancer (CRC) is the third most common cancer in men and the second in women worldwide,87 and around 30% of the patients will develop metastasis even after curative surgery It has been suggested that the HGF/MET axis is involved in the metastatic progression and potential invasiveness of the cancer cells by regulating the expression of cadherins and extracellular membrane proteases.88 Besides, MET amplification seems to be a late event in CRC progression MET amplification is more common in advanced tumour stages89 and its expression has been found to be higher in metastatic tissue than in primary tumour.90 Although survival of patients with unresectable metastatic CRC (mCRC) has improved in the last years with the introduction of agents targeting EGFR, such as cetuximab, its response rate range varies from 10% to 20%.91 92 HGF-induced MET activation has been described as a novel mechanism for cetuximab resistance Dual activation of both EGFR and MET receptors in CRC cells synergistically increases cell proliferation.93 Cetuximab could inhibit this cell growth by 60%–80% However, addition of HGF to cetuximab-treated cells phosphorylated MET, but not EGFR, restoring cell proliferation Inhibition of the HGF/MET axis may therefore improve response to EGFR inhibitors in CRC, and combination therapy should be further investigated.93 A recent study revealed that high levels of serum HGF and epiregulin (EREG) before treatment with anti-EGFR antibodies were associated with poor survival in KRAS (Kirsten rat sarcoma viral oncogene homolog) wild type patients with mCRC, suggesting that serum HGF and EREG may be associated with resistance to anti-EGFR treatment HGF might be a potential biomarker for predicting response and prognosis in dual target therapy with anti-EGFR antibodies and HGF/MET inhibitors.94 Handling editor Runjan Chetty Contributors Study concept and design: ABF-P and AEF; literature review, analysis and interpretation of literature, drafting of the manuscript: MMP and ABF-P; revision of manuscript, supervision of work: AEF and LRJ Competing interests None declared Provenance and peer review Commissioned; internally peer reviewed REFERENCES 10 11 12 13 Take home messages 14 ▸ HGF is a multifunctional cytokine produced by cells of mesenchymal origin and is involved in cell proliferation, survival, motility and morphogenesis ▸ Mesenchymal-epithelial transition (MET) is a tyrosine kinase receptor physiologically expressed on cells of epithelial origin and HGF is its unique binding factor ▸ The HGF/MET axis plays important roles in embryogenesis, organogenesis, adult tissue regeneration and carcinogenesis ▸ Both HGF and MET genes are overexpressed, mutated, and/ or amplified in several types of cancers, including haematological malignancies ▸ The HGF/MET axis has become the candidate target of numerous therapeutic clinical trials 15 16 17 18 19 20 Fajardo-Puerta AB, et al J Clin Pathol 2016;69:575–579 doi:10.1136/jclinpath-2015-203575 Matsumoto K, Nakamura T Hepatocyte growth factor: molecular structure, roles in liver regeneration, and other biological functions Crit Rev Oncog 1992;3:27–54 Trusolino L, Bertotti A, Comoglio PM MET signalling: principles and functions in development, organ regeneration and cancer Nat Rev Mol Cell Biol 2010;11:834–48 Eder JP, Vande Woude GF, Boerner SA, et al Novel therapeutic inhibitors of the c-Met signaling pathway in cancer Clin Cancer Res 2009;15:2207–14 Sattler M, Salgia R c-Met and hepatocyte growth factor: potential as novel targets in cancer therapy Curr Oncol Rep 2007;9:102–8 Comoglio PM, Giordano S, Trusolino L Drug development of MET inhibitors: targeting oncogene addiction and expedience Nat Rev Drug Discov 2008;7:504–16 Nakamura T, Sakai K, Nakamura T, et al Hepatocyte growth factor twenty years on: much more than a growth factor J Gastroenterol Hepatol 2011;26(Suppl 1):188–202 Barrow-McGee R, Kermorgant S Met endosomal signalling: in the right place, at the right time Int J Biochem Cell Biol 2014;49:69–74 Matsumoto K, Funakoshi H, Takahashi H, et al HGF-Met pathway in regeneration and drug discovery Biomedicines 2014;2:275–300 Zarnegar R, Petersen B, DeFrances MC, et al Localization of hepatocyte growth factor (HGF) gene on human chromosome Genomics 1992;12:147–50 Fukuyama R, Ichijoh Y, Minoshima S, et al Regional localization of the hepatocyte growth factor (HGF) gene to human chromosome band q21.1 Genomics 1991;11:410–15 Seki T, Hagiya M, Shimonishi M, et al Organization of the human hepatocyte growth factor-encoding gene Gene 1991;102:213–19 Schultz JM, Khan SN, Ahmed ZM, et al Noncoding mutations of HGF are associated with nonsyndromic hearing loss, DFNB39 Am J Hum Genet 2009;85:25–39 Jiang W, Hiscox S, Matsumoto K, et al Hepatocyte growth factor/scatter factor, its molecular, cellular and clinical implications in cancer Crit Rev Oncol Hematol 1999;29:209–48 Itoh H, Naganuma S, Takeda N, et al Regeneration of injured intestinal mucosa is impaired in hepatocyte growth factor activator-deficient mice Gastroenterology 2004;127:1423–35 Marchand-Adam S, Fabre A, Mailleux AA, et al Defect of pro-hepatocyte growth factor activation by fibroblasts in idiopathic pulmonary fibrosis Am J Respir Crit Care Med 2006;174:58–66 Zarneger R, Defrances MC, Michalopoulos GK Chapter 43 Hepatocyte growth factor: its role in hepatic growth and pathobiology The liver: biology and pathobiology 5th edn 2009:611–29 Bottaro DP, Rubin JS, Faletto DL, et al Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product Science 1991;251:802–4 Naldini L, Vigna E, Narsimhan RP, et al Hepatocyte growth factor (HGF) stimulates the tyrosine kinase activity of the receptor encoded by the proto-oncogene c-MET Oncogene 1991;6:501–4 Dean M, Park M, Le Beau MM, et al The human met oncogene is related to the tyrosine kinase oncogenes Nature 1985;318:385–8 Matsumoto K, Kataoka H, Date K, et al Cooperative interaction between alphaand beta-chains of hepatocyte growth factor on c-Met receptor confers 577 Gene of the month 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 578 ligand-induced receptor tyrosine phosphorylation and multiple biological responses J Biol Chem 1998;273:22913–20 Hartmann G, Naldini L, Weidner KM, et al A functional domain in the heavy chain of scatter factor/hepatocyte growth factor binds the c-Met receptor and induces cell dissociation but not mitogenesis Proc Natl Acad Sci USA 1992;89: 11574–8 Ponzetto C, Bardelli A, Maina F, et al A novel recognition motif for phosphatidylinositol 3-kinase binding mediates its association with the hepatocyte growth factor/scatter factor receptor Mol Cell Biol 1993;13:4600–8 Ponzetto C, Bardelli A, Zhen Z, et al A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family Cell 1994;77:261–71 Weidner KM, Di Cesare S, Sachs M, et al Interaction between Gab1 and the c-Met receptor tyrosine kinase is responsible for epithelial morphogenesis Nature 1996;384:173–6 Fixman ED, Fournier TM, Kamikura DM, et al Pathways downstream of Shc and Grb2 are required for cell transformation by the tpr-Met oncoprotein J Biol Chem 1996;271:13116–22 Boccaccio C, Andò M, Tamagnone L, et al Induction of epithelial tubules by growth factor HGF depends on the STAT pathway Nature 1998;391:285–8 Sipeki S, Bander E, Buday L, et al Phosphatidylinositol 3-kinase contributes to Erk1/ Erk2 MAP kinase activation associated with hepatocyte growth factor-induced cell scattering Cell Signal 1999;11:885–90 Maroun CR, Holgado-Madruga M, Royal I, et al The Gab1 PH domain is required for localization of Gab1 at sites of cell-cell contact and epithelial morphogenesis downstream from the met receptor tyrosine kinase Mol Cell Biol 1999;19:1784–99 Maroun CR, Naujokas MA, Holgado-Madruga M, et al The tyrosine phosphatase SHP-2 is required for sustained activation of extracellular signal-regulated kinase and epithelial morphogenesis downstream from the met receptor tyrosine kinase Mol Cell Biol 2000;20:8513–25 Muller M, Morotti A, Ponzetto C Activation of NF-kappaB is essential for hepatocyte growth factor-mediated proliferation and tubulogenesis Mol Cell Biol 2002;22:1060–72 Son BR, Marquez-Curtis LA, Kucia M, et al Migration of bone marrow and cord blood mesenchymal stem cells in vitro is regulated by stromal-derived factor-1-CXCR4 and hepatocyte growth factor-c-met axes and involves matrix metalloproteinases Stem Cells 2006;24:1254–64 Tulasne D, Deheuninck J, Lourenco FC, et al Proapoptotic function of the MET tyrosine kinase receptor through caspase cleavage Mol Cell Biol 2004;24:10328–39 Toschi L, Jänne PA Single-agent and combination therapeutic strategies to inhibit hepatocyte growth factor/MET signaling in cancer Clin Cancer Res 2008;14:5941–6 Peschard P, Park M Escape from Cbl-mediated downregulation: a recurrent theme for oncogenic deregulation of receptor tyrosine kinases Cancer Cell 2003;3: 519–23 Boissinot M, Vilaine M, Hermouet S The Hepatocyte Growth Factor (HGF)/met axis: a neglected target in the treatment of chronic myeloproliferative neoplasms? Cancers (Basel) 2014;6:1631–69 Russell WE, McGowan JA, Bucher NL Partial characterization of a hepatocyte growth factor from rat platelets J Cell Physiol 1984;119:183–92 Nakamura T, Nawa K, Ichihara A Partial purification and characterization of hepatocyte growth factor from serum of hepatectomized rats Biochem Biophys Res Commun 1984;122:1450–9 Nakamura T, Nawa K, Ichihara A, et al Purification and subunit structure of hepatocyte growth factor from rat platelets FEBS Lett 1987;224:311–16 Gohda E, Tsubouchi H, Nakayama H, et al Purification and partial characterization of hepatocyte growth factor from plasma of a patient with fulminant hepatic failure J Clin Invest 1988;81:414–19 Stoker M, Gherardi E, Perryman M, et al Scatter factor is a fibroblast-derived modulator of epithelial cell mobility Nature 1987;327:239–42 Gherardi E, Gray J, Stoker M, et al Purification of scatter factor, a fibroblast-derived basic protein that modulates epithelial interactions and movement Proc Natl Acad Sci USA 1989;86:5844–8 Weidner KM, Arakaki N, Hartmann G, et al Evidence for the identity of human scatter factor and human hepatocyte growth factor Proc Natl Acad Sci USA 1991;88:7001–5 Weidner KM, Behrens J, Vandekerckhove J, et al Scatter factor: molecular characteristics and effect on the invasiveness of epithelial cells J Cell Biol 1990;111 (Pt 1):2097–108 Naldini L, Weidner KM, Vigna E, et al Scatter factor and hepatocyte growth factor are indistinguishable ligands for the MET receptor EMBO J 1991;10:2867–78 Shima N, Nagao M, Ogaki F, et al Tumor cytotoxic factor/hepatocyte growth factor from human fibroblasts: cloning of its cDNA, purification and characterization of recombinant protein Biochem Biophys Res Commun 1991;180:1151–8 Uehara Y, Minowa O, Mori C, et al Placental defect and embryonic lethality in mice lacking hepatocyte growth factor/scatter factor Nature 1995;373:702–5 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 Schmidt C, Bladt F, Goedecke S, et al Scatter factor/hepatocyte growth factor is essential for liver development Nature 1995;373:699–702 Maroun CR, Rowlands T The Met receptor tyrosine kinase: a key player in oncogenesis and drug resistance Pharmacol Ther 2014;142:316–38 Zhuang Z, Park WS, Pack S, et al Trisomy 7-harbouring non-random duplication of the mutant MET allele in hereditary papillary renal carcinomas Nat Genet 1998;20:66–9 Cecchi F, Rabe DC, Bottaro DP Targeting the HGF/Met signaling pathway in cancer therapy Expert Opin Ther Targets 2012;16:553–72 Michalopoulos GK Advances in liver regeneration Expert Rev Gastroenterol Hepatol 2014;8:897–907 Mohammed FF, Khokha R Thinking outside the cell: proteases regulate hepatocyte division Trends Cell Biol 2005;15:555–63 Lindroos PM, Zarnegar R, Michalopoulos GK Hepatocyte growth factor (hepatopoietin A) rapidly increases in plasma before DNA synthesis and liver regeneration stimulated by partial hepatectomy and carbon tetrachloride administration Hepatology 1991;13:743–50 Michalopoulos GK Liver regeneration J Cell Physiol 2007;213:286–300 Kinoshita T, Hirao S, Matsumoto K, et al Possible endocrine control by hepatocyte growth factor of liver regeneration after partial hepatectomy Biochem Biophys Res Commun 1991;177:330–5 Yanagita K, Nagaike M, Ishibashi H, et al Lung May have an endocrine function producing hepatocyte growth factor in response to injury of distal organs Biochem Biophys Res Commun 1992;182:802–9 Ferlay J, Shin HR, Bray F, et al Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008 Int J Cancer 2010;127:2893–917 Marquardt JU, Galle PR, Teufel A Molecular diagnosis and therapy of hepatocellular carcinoma (HCC): an emerging field for advanced technologies J Hepatol 2012;56:267–75 Whittaker S, Marais R, Zhu AX The role of signaling pathways in the development and treatment of hepatocellular carcinoma Oncogene 2010;29:4989–5005 Venepalli NK, Goff L Targeting the HGF-cMET axis in hepatocellular carcinoma Int J Hepatol 2013;2013:341636 Vejchapipat P, Tangkijvanich P, Theamboonlers A, et al Association between serum hepatocyte growth factor and survival in untreated hepatocellular carcinoma J Gastroenterol 2004;39:1182–8 Kiss A, Wang NJ, Xie JP, et al Analysis of transforming growth factor (TGF)-alpha/ epidermal growth factor receptor, hepatocyte growth Factor/c-met,TGF-beta receptor type II, and p53 expression in human hepatocellular carcinomas Clin Cancer Res 1997;3:1059–66 Boix L, Rosa JL, Ventura F, et al c-met mRNA overexpression in human hepatocellular carcinoma Hepatology 1994;19:88–91 Osada S, Kanematsu M, Imai H, et al Clinical significance of serum HGF and c-Met expression in tumor tissue for evaluation of properties and treatment of hepatocellular carcinoma Hepatogastroenterology 2008;55:544–9 Suzuki K, Hayashi N, Yamada Y, et al Expression of the c-met protooncogene in human hepatocellular carcinoma Hepatology 1994;20:1231–6 Ueki T, Fujimoto J, Suzuki T, et al Expression of hepatocyte growth factor and its receptor c-met proto-oncogene in hepatocellular carcinoma Hepatology 1997;25:862–6 Kaposi-Novak P, Lee JS, Gòmez-Quiroz L, et al Met-regulated expression signature defines a subset of human hepatocellular carcinomas with poor prognosis and aggressive phenotype J Clin Invest 2006;116:1582–95 Okano J, Shiota G, Kawasaki H Expression of hepatocyte growth factor (HGF) and HGF receptor (c-met) proteins in liver diseases: an immunohistochemical study Liver 1999;19:151–9 Junbo H, Li Q, Zaide W, et al Increased level of serum hepatocyte growth factor/ scatter factor in liver cancer is associated with tumor metastasis In Vivo 1999;13:177–80 Daveau M, Scotte M, Franỗois A, et al Hepatocyte growth factor, transforming growth factor alpha, and their receptors as combined markers of prognosis in hepatocellular carcinoma Mol Carcinog 2003;36:130–41 Wu FS, Zheng SS, Wu LJ, et al Study on the prognostic value of hepatocyte growth factor and c-met for patients with hepatocellular carcinoma Zhonghua Wai Ke Za Zhi 2006;44:603–8 Ke AW, Shi GM, Zhou J, et al Role of overexpression of CD151 and/or c-Met in predicting prognosis of hepatocellular carcinoma Hepatology 2009;49:491–503 Wang ZL, Liang P, Dong BW, et al Prognostic factors and recurrence of small hepatocellular carcinoma after hepatic resection or microwave ablation: a retrospective study J Gastrointest Surg 2008;12:327–37 Santoro A, Rimassa L, Borbath I, et al Tivantinib for second-line treatment of advanced hepatocellular carcinoma: a randomised, placebo-controlled phase study Lancet Oncol 2013;14:55–63 Santoro A, Simonelli M, Rodriguez-Lope C, et al A Phase-1b study of tivantinib (ARQ 197) in adult patients with hepatocellular carcinoma and cirrhosis Br J Cancer 2013;108:21–4 Takeda S, Liu H, Sasagawa S, et al HGF-MET signals via the MLL-ETS2 complex in hepatocellular carcinoma J Clin Invest 2013;123:3154–65 Fajardo-Puerta AB, et al J Clin Pathol 2016;69:575–579 doi:10.1136/jclinpath-2015-203575 Gene of the month 77 78 79 80 81 82 83 84 85 Hack SP, Bruey JM, Koeppen H HGF/MET-directed therapeutics in gastroesophageal cancer: a review of clinical and biomarker development Oncotarget 2014;5:2866–80 Liu YJ, Shen D, Yin X, et al HER2, MET and FGFR2 oncogenic driver alterations define distinct molecular segments for targeted therapies in gastric carcinoma Br J Cancer 2014;110:1169–78 Nagatsuma AK, Aizawa M, Kuwata T, et al Expression profiles of HER2, EGFR, MET and FGFR2 in a large cohort of patients with gastric adenocarcinoma Gastric Cancer 2015;18:227–38 Lee JH, Han SU, Cho H, et al A novel germ line juxtamembrane Met mutation in human gastric cancer Oncogene 2000;19:4947–53 Chong IY, Cunningham D, Barber LJ, et al The genomic landscape of oesophagogastric junctional adenocarcinoma J Pathol 2013;231:301–10 Lordick F, Allum W, Carneiro F, et al Unmet needs and challenges in gastric cancer: the way forward Cancer Treat Rev 2014;40:692–700 Oliner KS, Tang R, Anderson A, et al Evaluation of MET pathway biomarkers in a phase II study of rilotumumab (R, AMG 102) or placebo (P) in combination with epirubicin, cisplatin, and capecitabine (ECX) in patients ( pts) with locally advanced or metastatic gastric (G) or esophagogastric junction (EGJ) cancer J Clin Oncol 2012 (suppl; abstr 4005) Kawakami H, Okamoto I, Arao T, et al MET amplification as a potential therapeutic target in gastric cancer Oncotarget 2013;4:9–17 Lennerz JK, Kwak EL, Ackerman A, et al MET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinib J Clin Oncol 2011;29:4803–10 86 87 88 89 90 91 92 93 94 Fajardo-Puerta AB, et al J Clin Pathol 2016;69:575–579 doi:10.1136/jclinpath-2015-203575 Janjigian YY, Tang LH, Coit DG, et al MET expression and amplification in patients with localized gastric cancer Cancer Epidemiol Biomarkers Prev 2011;20:1021–7 World Health Organisation Globocan 2012: Estimated Cancer Incidence, Mortality and Prevalence Worldwide in 2012 Secondary Globocan 2012: Estimated Cancer Incidence, Mortality and Prevalence Worldwide in 2012 2012 http://globocan.iarc fr/Pages/fact_sheets_cancer.aspx Gao D, Vahdat LT, Wong S, et al Microenvironmental regulation of epithelial-mesenchymal transitions in cancer Cancer Res 2012;72:4883–9 Zeng ZS, Weiser MR, Kuntz E, et al c-Met gene amplification is associated with advanced stage colorectal cancer and liver metastases Cancer Lett 2008;265:258–69 Di Renzo MF, Olivero M, Giacomini A, et al Overexpression and amplification of the met/HGF receptor gene during the progression of colorectal cancer Clin Cancer Res 1995;1:147–54 Jonker DJ, O’Callaghan CJ, Karapetis CS, et al Cetuximab for the treatment of colorectal cancer N Engl J Med 2007;357:2040–8 Arnold D, Seufferlein T Targeted treatments in colorectal cancer: state of the art and future perspectives Gut 2010;59:838–58 Liska D, Chen CT, Bachleitner-Hofmann T, et al HGF rescues colorectal cancer cells from EGFR inhibition via MET activation Clin Cancer Res 2011;17:472–82 Takahashi N, Yamada Y, Furuta K, et al Serum levels of hepatocyte growth factor and epiregulin are associated with the prognosis on anti-EGFR antibody treatment in KRAS wild-type metastatic colorectal cancer Br J Cancer 2014;110:2716–27 579 ... further studies on the involvement of HGF as a modulator of cellular growth and motility during embryogenesis, tissue regeneration and carcinogenesis Interestingly, targeted disruption of HGF. .. cells of epithelial origin and HGF is its unique binding factor ▸ The HGF/ MET axis plays important roles in embryogenesis, organogenesis, adult tissue regeneration and carcinogenesis ▸ Both HGF. .. (HGF) stimulates the tyrosine kinase activity of the receptor encoded by the proto-oncogene c-MET Oncogene 1991;6:501–4 Dean M, Park M, Le Beau MM, et al The human met oncogene is related to the

Ngày đăng: 04/12/2022, 10:32

TÀI LIỆU CÙNG NGƯỜI DÙNG

TÀI LIỆU LIÊN QUAN