Jump to content

Hepatocyte growth factor receptor: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
Citation bot (talk | contribs)
m Citation maintenance. [Pu]Formatted: doi. You can use this bot yourself! Please report any bugs.
Line 134: Line 134:
* [http://www.expasy.org/uniprot/P08581 UniProtKB/Swiss-Prot entry P08581: MET_HUMAN], ExPASy (Expert Protein Analysis System) proteomics server of the Swiss Institute of Bioinformatics (SIB).
* [http://www.expasy.org/uniprot/P08581 UniProtKB/Swiss-Prot entry P08581: MET_HUMAN], ExPASy (Expert Protein Analysis System) proteomics server of the Swiss Institute of Bioinformatics (SIB).
* [[Tpr-met fusion protein]]
* [[Tpr-met fusion protein]]
* A [http://www.vai.org/met/ table] with references to significant roles of MET in cancer.


==References==
==References==

Revision as of 21:03, 11 June 2009

Template:PBB MET (mesenchymal-epithelial transition factor) is a proto-oncogene that encodes a protein MET, also known as c-Met or hepatocyte growth factor receptor (HGFR). MET is a membrane receptor that is essential for embryonic development and wound healing. Hepatocyte growth factor (HGF) is the only known ligand of the MET receptor. MET is normally expressed by cells of epithelial origin, while expression of HGF is restricted to cells of mesenchymal origin. Upon HGF stimulation, MET induces several biological responses that collectively give rise to a program known as invasive growth. Abnormal MET activation in cancer correlates with poor prognosis, where aberrantly active MET triggers tumor growth, formation of new blood vessels (angiogenesis) that supply the tumor with nutrients, and cancer spread to other organs (metastasis). MET is deregulated in many types of human malignancies, including cancers of kidney, liver, stomach, breast, and brain. Normally, only stem cells and progenitor cells express MET, which allows these cells to grow invasively in order to generate new tissues in an embryo or regenerate damaged tissues in an adult. However, cancer stem cells are thought to hijack the ability of normal stem cells to express MET, and thus become the cause of cancer persistence and spread to other sites in the body.

Template:PBB Summary

MET Gene

MET proto-oncogene (GeneID: 4233) has a total length of 125,982 bp, and it is located in the 7q31 locus of chromosome 7. MET is transcribed into a 6,641 bp mature mRNA, which is then translated into a 1,390 amino-acid MET protein.

MET Protein

Schematic structure of MET protein [1]

MET is a receptor tyrosine kinase (RTK) that is produced as a single-chain precursor. The precursor is proteolytically cleaved at a furin site to yield a highly glycosylated extracellular α-subunit and a transmembrane β-subunit, which are linked together by a disulfide bridge. [2]

Extracellular Portion:

  1. Region of homology to semaphorins (Sema domain), which includes the full α-chain and the N-terminal part of the β-chain;
  2. Cysteine-rich MET-related sequence (MRS domain);
  3. Glycine-proline-rich repeats (G-P repeats);
  4. Four immunoglobulin-like structures (Ig domains), a typical protein-protein interaction region. [2]


Intracellular Portion:

  1. Juxtamembrane segment that contains:
  2. Tyrosine kinase domain, which mediates MET biological activity. Following MET activation, transphosphorylation occurs on Tyr 1234 and Tyr 1235;
  3. C-terminal region contains two crucial tyrosines (Tyr 1349 and Tyr 1356), which are inserted into the multisubstrate docking site, capable of recruiting downstream adapter proteins with Src homology-2 (SH2) domains. [5] The two tyrosines of the docking site have been reported to be necessary and sufficient for the signal transduction both in vitro [6] and in vivo [7].

MET Signaling Pathway

MET signaling complex [8]

MET activation by its ligand HGF induces MET kinase catalytic activity, which triggers transphosphorylation of the tyrosines Tyr 1234 and Tyr 1235. These two tyrosines engage various signal transducers, thus initiating a whole spectrum of biological activities driven by MET, collectively knows as the invasive growth program. The transducers interact with the intracellular multisubstrate docking site of MET either directly, such as GRB2, SHC [9], SRC, and the p85 regulatory subunit of phosphatidylinositol-3 kinase (PI3K) [6], or indirectly through the scaffolding protein Gab1 [10]. Tyr 1349 and Tyr 1356 of the multisubstrate docking site are both involved in the interaction with GAB1, SRC, and SHC, while only Tyr 1356 is involved in the recruitment of GRB2, phospholipase C γ (PLC-γ), p85, and SHP2 [11]. GAB1 is a key coordinator of the cellular responses to MET and binds the MET intracellular region with high avidity, but low affinity[12]. Upon interaction with MET, GAB1 becomes phosphorylated on several tyrosine residues which, in turn, recruit a number of signalling effectors, including PI3K, SHP2, and PLC-γ. GAB1 phosphorylation by MET results in a sustained signal that mediates most of the downstream signaling pathways [13].

MET engagement activates multiple signal transduction pathways:

  1. The RAS pathway mediates HGF-induced scattering and proliferation signals, which lead to branching morphogenesis [14]. Of note, HGF, differently from most mitogens, induces sustained RAS activation, and thus prolonged MAPK activity [15].
  2. The PI3K pathway is activated in two ways: PI3K can be either downstream of RAS, or it can be recruited directly through the multifunctional docking site [16]. Activation of the PI3K pathway is currently associated with cell motility through remodeling of adhesion to the extracellular matrix as well as localized recruitment of transducers involved in cytoskeletal reorganization, such as RAC1 and PAK. PI3K activation also triggers a survival signal due to activation of the AKT pathway [1].
  3. The STAT pathway, together with the sustained MAPK activation, is necessary for the HGF-induced branching morphogenesis. MET activates the STAT3 transcription factor directly, through an SH2 domain [17].
  4. The beta catenin pathway, a key component of the Wnt signaling pathway, translocates into the nucleus following MET activation and participates in transcriptional regulation of numerous genes [18].
  5. The Notch pathway, through transcriptional activation of Delta ligand [8][19].
Interplay between MET, beta catenin, Wnt, and Notch signaling pathways [8]

Essential Role in Development

MET mediates a complex program known as invasive growth [1]. Activation of MET triggers mitogenesis, and morphogenesis [20]. During embryonic development, transformation of the flat, two-layer germinal disc into a three-dimensional body depends on transition of some cells from an epithelial phenotype to spindle-shaped cells with motile behaviour, a mesenchymal phenotype. This process is referred to as epithelial-mesenchymal transition (EMT). [8] Later in embryonic development, MET is crucial for gastrulation, angiogenesis, myoblast migration, bone remodeling, and nerve sprouting among others [21]. MET is essential for embryogenesis, because MET -/- mice die in utero due to severe defects in placental development [22]. Furthermore, MET is required for such critical processes as liver regeneration and wound healing during adulthood [1].

Expression of HGF and MET

Location

MET is normally expressed by epithelial cells [1]. However, MET is also found on endothelial cells, neurons, hepatocytes, hematopoietic cells, and melanocytes [23]. HGF expression is restricted to cells of mesenchymal origin [8].

Transcriptional Control

MET transcription is activated by HGF and several growth factors [24].

Ets1

MET promoter has four putative binding sites for Ets, a family of transcription factors that control several invasive growth genes [24]. Ets1 activates MET transcription in vitro [25].

HIF1

MET transcription is activated by hypoxia-inducible factor 1 (HIF1), which is activated by low concentration of intracellular oxygen [26]. HIF1 can bind to one of the several hypoxia response elements (HREs) in the MET promoter [8].

AP-1

Hypoxia also activates transcription factor AP-1, which is involved in MET transcription [8].

Central Role in Cancer

MET pathway plays an important role in the development of cancer through:

  1. activation of key oncogenic pathways (RAS, PI3K, STAT3, beta catenin);
  2. angiogenesis (sprouting of new blood vessels from pre-existing ones to supply a tumor with nutrients);
  3. scatter (cells dissociation due to metalloprotease production), which often leads to metastasis [27].

MET and Tumour Suppresor Genes

PTEN

PTEN (phosphatase and tensin homolog) is a tumor suppressor gene encoding a protein PTEN, which possesses lipid and protein phosphatase-dependent as well as phosphatase-independent activities [28]. PTEN protein phosphatase is able to interfere with MET signaling by dephosphorylating either PIP3 generated by PI3K, or the p52 isoform of SHC. SHC dephosphorylation inhibits recruitment of the GRB2 adapter to activated MET. [29]

VHL

There is evidence of correlation between inactivation of VHL tumor suppressor gene and increased MET signaling in renal cell carcinoma (RCC) [30].

Cancer Therapies Targeting HGF/MET

Strategies to inhibit biological activity of MET [1]

Since tumor invasion and metastasis are the main cause of death in cancer patients, interfering with MET signaling appears to be a promising therapeutic approach.

MET Kinase Inhibitors

Kinase inhibitors are low molecular weight molecules that prevent ATP binding to MET, thus inhibiting receptor transphosphorylation and recruitment of the downstream effectors. The limitations of kinase inhibitors include the facts that they only inhibit kinase-dependent MET activation, and that none of them is fully specific for MET.

  1. K252a (Fermentek Biotechnology) is a staurosporine analogue isolated from Nocardiopsis sp. soil fungi, and it is a potent inhibitor of all receptor tyrosine kinases (RTKs). At nanomolar concentrations, K252a inhibits both the wild type and the mutant (M1268T) MET function. [31]
  2. SU11274 (Sugen) specifically inhibits MET kinase activity and its subsequent signaling. SU11274 is also an effective inhibitor of the M1268T and H1112Y MET mutants, but not the L1213V and Y1248H mutants [32]. SU11274 has been demonstrated to inhibit HGF-induced motility and invasion of epithelial and carcinoma cells [33].
  3. PHA-665752 (Pfizer) specifically inhibits MET kinase activity, and it has been demonstrated to represses both HGF-dependent and constitutive MET phosphorylation [34]. Furthermore, some tumors harboring MET amplifications are highly sensitive to treatment with PHA-665752 [35].
  4. ARQ197 (ArQule) is a promising selective inhibitor of MET, which has entered a phase 2 clinical trial in 2008.
  5. XL880 (Exelixis) targets multiple receptor tyrosine kinases (RTKs) with growth-promoting and angiogenic properties. The primary targets of XL880 are MET, VEGFR2, and KDR. XL880 has completed a phase 2 clinical trials with indications for papillary renal cell carcinoma, gastric cancer, and head and neck cancer (see Poster).
  6. SGX523 (SGX Pharmaceuticals) specifically inhibits MET at low nanomolar concentrations. (See Poster.)
  7. MP470 (SuperGen) is a novel inhibitor of c-KIT, MET, PDGFR, Flt3, and AXL. Phase I clinical trial of MP470 had been announced in 2007. (See Poster.)

HGF Inhibitors

Since HGF is the only known ligand of MET, formation of a HGF:MET complex blocks MET biological activity. For this purpose, truncated HGF, anti-HGF neutralizing antibodies, and an uncleavable form of HGF have been utilized so far. The major limitation of HGF inhibitors is that they block only HGF-dependent MET activation.

  1. NK4 competes with HGF as it binds MET without inducing receptor activation, thus behaving as a full antagonist. NK4 is a molecule bearing the N-terminal hairpin and the four kringle domains of HGF. Moreover, NK4 is structurally similar to angiostatins, which is why it possesses anti-angiogenic activity. [36]
  2. Neutralizing anti-HGF antibodies were initially tested in combination, and it was shown that at least three antibodies, acting on different HGF epitopes, are necessary to prevent MET tyrosine kinase activation [37]. More recently, it has been demonstrated that fully human monoclonal antibodies can individually bind and neutralize human HGF, leading to regression of tumors in mouse models [38]. Two anti-HGF antibodies are currently available: the humanized AV299 (AVEO), and the fully human AMG102 (Amgen).
  3. Uncleavable HGF is an engineered form of pro-HGF carrying a single amino-acid substitution, which prevents the maturation of the molecule. Uncleavable HGF is capable of blocking MET-induced biological responses by binding MET with high affinity and displacing mature HGF. Moreover, uncleavable HGF competes with the wild-type endogenous pro-HGF for the catalytic domain of proteases that cleave HGF precursors. Local and systemic expression of uncleavable HGF inhibits tumor growth and, more importantly, prevents metastasis. [39]

Decoy MET

Decoy MET refers to a soluble truncated MET receptor. Decoys are able to inhibit MET activation mediated by both HGF-dependent and independent mechanisms, as decoys prevent both the ligand binding and the MET receptor homodimerization. CGEN241 (Compugen) is a decoy MET that is highly efficient in inhibiting tumor growth and preventing metastasis in animal models. [40]

Immunotherapy Targeting MET

Drugs used for immunotherapy can act either passively by enhancing the immunologic response to MET-expressing tumor cells, or actively by stimulating immune cells and altering differentiation/growth of tumor cells [41].

  • Passive Immunotherapy
Administering monoclonal antibodies (mAbs) is a form of passive immunotherapy. MAbs facilitate destruction of tumor cells by complement-dependent cytotoxicity (CDC) and cell-mediated cytotoxicity (ADCC). In CDC, mAbs bind to specific antigen, leading to activation of the complement cascade, which in turn leads to formation of pores in tumor cells. In ADCC, the Fab domain of a mAb binds to a tumor antigen, and Fc domain binds to Fc receptors present on effector cells (phagocytes and NK cells), thus forming a bridge between an effector and a target cells. This induces the effector cell activation, leading to phagocytosis of the tumor cell by neutrophils and macrophages. Furthermore, NK cells release cytotoxic molecules, which lyse tumor cells. [41]
  1. DN30 is monoclonal anti-MET antibody that recognizes the extracellular portion of MET. DN30 induces both shedding of the MET ectodomain as well as cleavage of the intracellular domain, which is successively degraded by proteasome machinery. As a consequence, on one side MET is inactivated, and on the other side the shed portion of extracellular MET hampers activation of other MET receptors, acting as a decoy. DN30 inhibits tumour growth and prevents metastasis in animal models. [42]
  2. OA-5D5 is one-armed monoclonal anti-MET antibody that was demonstrated to inhibit orthotopic pancreatic [43] and glioblastoma [44] tumor growth and to improve survival in tumor xenograft models. OA-5D5 is produced as a recombinant protein in Escherichia coli. It is composed of murine variable domains for the heavy and light chains with human IgG1 constant domains. The antibody blocks HGF binding to MET in a competitive fashion.
  • Active Immunotherapy
Active immunotherapy to MET-expressing tumors can be achieved by administering cytokines, such as interferons (IFNs) and interleukins (IL-2), which triggers non-specific stimulation of numerous immune cells. IFNs have been tested as therapies for many types of cancers and have demonstrated therapeutic benefits. IL-2 has been approved by FDA for the treatment of renal cell carcinoma and metastatic melanoma, which often have deregulated MET activity. [41]

References

  1. ^ a b c d e f Gentile A, Trusolino L, Comoglio PM. (2008). The Met tyrosine kinase receptor in development and cancer. Cancer Metastasis Reviews, 27(1):85-94. PMID: 18175071
  2. ^ a b Birchmeier, C., Birchmeier, W., Gherardi, E., & Vande Woude, G. F. (2003). Met, metastasis, motility and more. Nature Reviews. Molecular Cell Biology, 4: 915–925. PMID: 14685170
  3. ^ Gandino L, Longati P, Medico E, Prat M, Comoglio PM. 1994. Phosphorylation of serine 985 negatively regulates the hepatocyte growth factor receptor kinase. J Biol Chem, 269(3):1815-20. PMID: 8294430
  4. ^ Peschard P, Fournier TM, Lamorte L, Naujokas MA, Band H, Langdon WY, Park M. (2001). Mutation of the c-Cbl TKB domain binding site on the Met receptor tyrosine kinase converts it into a transforming protein. Mol Cell, 8(5): 995-1004. PMID: 11741535
  5. ^ Ponzetto C, Bardelli A, Zhen Z, Maina F, dalla Zonca P, Giordano S, Graziani A, Panayotou G, Comoglio PM. (1994). A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family. Cell, 77: 261-271. PMID 7513258
  6. ^ a b Maina F, Casagranda F, Audero E, Simeone A, Comoglio PM, Klein R, Ponzetto C. (1996). Uncoupling of Grb2 from the Met receptor in vivo reveals complex roles in muscle development. Cell, 87: 531–542. 8898205
  7. ^ Ponzetto C, Bardelli A, Zhen Z, Maina F, dalla Zonca P, Giordano S, Graziani A, Panayotou G, Comoglio PM. (1994). A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family. Cell, 77: 261–271. PMID: 7513258
  8. ^ a b c d e f g Boccaccio C, Comoglio PM. (2006). Invasive growth: a MET-driven genetic programme for cancer and stem cells. Nat Rev Cancer, 6(8): 637-45. PMID: 16862193
  9. ^ Pelicci G, Giordano S, Zhen Z, Salcini AE, Lanfrancone L, Bardelli A, Panayotou G, Waterfield MD, Ponzetto C, Pelicci PG, et al. (1995). The motogenic and mitogenic responses to HGF are amplified by the Shc adaptor protein. Oncogene, 10: 1631–1638. PMID: 7731718
  10. ^ Weidner KM, Di Cesare S, Sachs M, Brinkmann V, Behrens J, Birchmeier W. (1996). Interaction between Gab1 and the c-Met receptor tyrosine kinase is responsible for epithelial morphogenesis. Nature, 384: 173–176. PMID: 8906793
  11. ^ Furge KA, Zhang YW, Vande Woude GF. (2000). Met receptor tyrosine kinase: Enhanced signaling through adapter proteins. Oncogene, 19: 5582–5589. PMID: 11114738
  12. ^ Gual P, Giordano S, Anguissola S, Parker PJ, Comoglio PM. (2001). Gab1 phosphorylation: A novel mechanism for negative regulation of HGF receptor signaling. Oncogene, 20: 156–166. PMID: 11313945
  13. ^ Gual P, Giordano S, Williams TA, Rocchi S, Van Obberghen E, Comoglio PM. (2000). Sustained recruitment of phospholipase C-gamma to Gab1 is required for HGF-induced branching tubulogenesis. Oncogene, 19: 1509–1518. PMID: 10734310
  14. ^ O'Brien LE, Tang K, Kats ES, Schutz-Geschwender A, Lipschutz JH, Mostov KE. (2004). ERK and MMPs sequentially regulate distinct stages of epithelial tubule development. Developments in Cell, 7: 21–32. PMID: 15239951
  15. ^ Marshall, C J (1995). Specificity of receptor tyrosine kinase signaling: Transient versus sustained extracellular signal-regulated kinase activation. Cell, 80: 179–185. PMID: 7834738
  16. ^ Graziani A, Gramaglia D, Cantley LC, Comoglio PM.. (1991). The tyrosine-phosphorylated hepatocyte growth factor/scatter factor receptor associates with phosphatidylinositol 3-kinase. Journal of Biological Chemistry, 266, 22087–22090. PMID: 1718989
  17. ^ Boccaccio C, Andò M, Tamagnone L, Bardelli A, Michieli P, Battistini C, Comoglio PM. (1998). Induction of epithelial tubules by growth factor HGF depends on the STAT pathway. Nature, 391: 285–288. PMID: 9440692
  18. ^ Health Value: Interplay between Wnt and MET signaling pathways.
  19. ^ Gude NA, Emmanuel G, Wu W, Cottage CT, Fischer K, Quijada P, Muraski JA, Alvarez R, Rubio M, Schaefer E, Sussman MA. (2008). Activation of Notch-mediated protective signaling in the myocardium. Circulation Research, 102(9): 1025-35. PMID: 18369158
  20. ^ Health Value: Biological activity of MET
  21. ^ Birchmeier C and Gherardi E. (1998). Developmental roles of HGF/SF and its receptor, the c-Met tyrosine kinase. Trends in Cell Biology, 8(10):404-10. PMID: 9789329
  22. ^ Uehara Y, Minowa O, Mori C, Shiota K, Kuno J, Noda T, Kitamura N. (1995). Placental defect and embryonic lethality in mice lacking hepatocyte growth factor/scatter factor. Nature, 373: 702–705. PMID: 7854453
  23. ^ Health Value: MET expression.
  24. ^ a b Shirasaki F, Makhluf HA, LeRoy C, Watson DK, Trojanowska M. (1999). Ets transcription factors cooperate with Sp1 to activate the human tenascin-C promoter. Oncogene, 18: 7755–7764. PMID: 10618716
  25. ^ Gambarotta G, Boccaccio C, Giordano S, Andŏ M, Stella MC, Comoglio PM. (1996). Ets up-regulates MET transcription. Oncogene, 13: 1911–1917. PMID: 8934537
  26. ^ Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano S, Comoglio PM. (2003). Hypoxia promotes invasive growth by transcriptional activation of the Met protooncogene. Cancer Cell, 3, 347–361. PMID: 12726861
  27. ^ Health Value: Role of MET in cancer.
  28. ^ Leslie NR, Bennett D, Gray A, Pass I, Hoang-Xuan K and Downes CP. (2001). Biochem. J., 357, 427–435. PMID: 9593664
  29. ^ Abounader R, Reznik T, Colantuoni C, Martinez-Murillo F, Rosen EM, Laterra J. (2004). Regulation of c-Met-dependent gene expression by PTEN. Oncogene, 23(57): 9173-82. PMID: 15516982
  30. ^ Morris MR, Gentle D, Abdulrahman M, Maina EN, Gupta K, Banks RE, Wiesener MS, Kishida T, Yao M, Teh B, Latif F, Maher ER. (2005). Tumor suppressor activity and epigenetic inactivation of hepatocyte growth factor activator inhibitor type 2/SPINT2 in papillary and clear cell renal cell carcinoma. Cancer Research, 65(11): 4598-606. PMID: 15930277
  31. ^ Morotti A, Mila S, Accornero P, Tagliabue E, Ponzetto C. (2002). K252a inhibits the oncogenic properties of Met, the HGF receptor. Oncogene, 21: 4885–4893. PMID: 12118367
  32. ^ Berthou S, Aebersold DM, Schmidt LS, Stroka D, Heigl C, Streit B, Stalder D, Gruber G, Liang C, Howlett AR, Candinas D, Greiner RH, Lipson KE, Zimmer Y. (2004). The Met kinase inhibitor SU11274 exhibits a selective inhibition pattern toward different receptor mutated variants. Oncogene, 23, 5387–5393. PMID: 15064724
  33. ^ Xueyan Wang, Phuong Le, Congxin Liang, Julie Chan, David Kiewlich, Todd Miller, Dave Harris, Li Sun, Audie Rice, Stefan Vasile, Robert A. Blake, Anthony R. Howlett, Neela Patel, Gerald McMahon and Kenneth E. Lipson. (2003). Potent and selective inhibitors of the Met [hepatocyte growth factor/scatter factor (HGF/SF) receptor] tyrosine kinase block HGF/SF-induced tumor cell growth and invasion. Molecular Cancer Therapy, 2: 1085-1092. PMID: 14617781
  34. ^ Christensen JG, Schreck R, Burrows J, Kuruganti P, Chan E, Le P, Chen J, Wang X, Ruslim L, Blake R, Lipson KE, Ramphal J, Do S, Cui JJ, Cherrington JM, Mendel DB. (2003). A selective small molecule inhibitor of c-Met kinase inhibits c-Met-dependent phenotypes in vitro and exhibits cytoreductive antitumor activity in vivo. Cancer Research, 63: 7345–7355. PMID: 14612533
  35. ^ Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald H, Kim WJ, Okimoto RA, Bell DW, Sgroi DC, Christensen JG, Settleman J, Haber DA. (2006). Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proceedings of the National Academy of Sciences of the United States of America, 103, 2316–2321. PMID: 16461907
  36. ^ Matsumoto K, Nakamura T. (2003). NK4 (HGF-antagonist/angiogenesis inhibitor) in cancer biology and therapeutics. Cancer Science, 94: 321–327. PMID: 12824898
  37. ^ Cao B, Su Y, Oskarsson M, Zhao P, Kort EJ, Fisher RJ, Wang LM, Vande Woude GF. (2001). Neutralizing monoclonal antibodies to HGF show antitumor activity in xenografts. Proceedings of the National Academy of Sciences of the United States of America, 98: 7443–7448. PMID: 11416216
  38. ^ Burgess T, Coxon A, Meyer S, Sun J, Rex K, Tsuruda T, Chen Q, Ho SY, Li L, Kaufman S, McDorman K, Cattley RC, Sun J, Elliott G, Zhang K, Feng X, Jia XC, Green L, Radinsky R, Kendall R. (2006). Fully human monoclonal antibodies to hepatocyte growth factor with therapeutic potential against hepatocyte growth factor/c-Met-dependent human tumors. Cancer Research, 66: 1721–1729. PMID: 16452232
  39. ^ Mazzone M, Basilico C, Cavassa S, Pennacchietti S, Risio M, Naldini L, Comoglio PM, Michieli P. (2004). An uncleavable form of pro-scatter factor suppresses tumor growth and dissemination in mice. Journal of Clinical Investigation, 114: 1418–1432. PMID: 15545993
  40. ^ Michieli P, Mazzone M, Basilico C, Cavassa S, Sottile A, Naldini L, Comoglio PM. (2004). Targeting the tumor and its microenvironment by a dual-function decoy Met receptor. Cancer Cell, 6: 61–73. PMID: 15261142
  41. ^ a b c Reang P, Gupta M, Kohli K. (2006). Biological response modifiers in cancer. Medscape General Medicine, 8(4): 33. PMID: 17415315
  42. ^ Petrelli A, Circosta P, Granziero L, Mazzone M, Pisacane A, Fenoglio S, Comoglio PM, Giordano S. (2006). Ab-induced ectodomain shedding mediates hepatocyte growth factor receptor down-regulation and hampers biological activity. Proceedings of the National Academy of Sciences of the United States of America, 103: 5090–5095. PMID: 16547140
  43. ^ Jin H, Yang R, Zheng Z, Romero M, Ross J, Bou-Reslan H, Carano RA, Kasman I, Mai E, Young J, Zha J, Zhang Z, Ross S, Schwall R, Colbern G, Merchant M. (2008). MetMAb, the One-Armed 5D5 Anti-c-Met Antibody, Inhibits Orthotopic Pancreatic Tumor Growth and Improves Survival. Cancer Research, 68(11): 4360-4368. PMID: 18519697
  44. ^ Martens T, Schmidt NO, Eckerich C, Fillbrandt R, Merchant M, Schwall R, Westphal M, Lamszus K. (2006). A novel one-armed anti-c-Met antibody inhibits glioblastoma growth in vivo. Clinical Cancer Research, 12(20): 6144-52. PMID: 17062691

Further reading

  • Peruzzi B, Bottaro DP (2006). "Targeting the c-Met signaling pathway in cancer". Clin. Cancer Res. 12 (12): 3657–60. doi:10.1158/1078-0432.CCR-06-0818. PMID 16778093.
  • Birchmeier, C., Birchmeier, W., Gheradi, E., & Vande Woude, G. F. (2003). Met, metastasis, motility and more. Nature Reviews Molecular Cell Biology, 4, 915—925. PMID 14685170 doi:10.1038/nrm1261
  • Zhang, Y., & Vande Woude, G. F. (2003). HGF/SF-Met signaling in the control of branching morphogenesis and invasion. Journal of Cellular Biochemistry, 88, 408—417. PMID 12520544 doi:10.1002/jcb.10358
  • Paumelle, R., Tulashe, D., Kherrouche, Z., Plaza, S., Leroy, C., Reveneau, S., Vandenbunder, B., & Fafeur, V. (2002). Hepatocyte growth factor/scatter factor activates the ETS1 transcription factor by a RAS-RAF-MEK-ERK signaling pathway. Oncogene, 21, 2309—2319. PMID 11948414 doi:10.1038/sj.onc.1205297

Template:PBB Further reading

Template:PBB Controls