24 results on '"Selma Pennacchietti"'
Search Results
2. Supplementary Figure 2 from Tivantinib (ARQ197) Displays Cytotoxic Activity That Is Independent of Its Ability to Bind MET
- Author
-
Paolo Michieli, Guido Serini, Donatella Valdembri, Alberto Bardelli, Sabrina Arena, Cristina Chiriaco, Elisa Vigna, Selma Pennacchietti, and Cristina Basilico
- Abstract
PDF File - 269K, Tivantinib does not inhibit HGF-dependent or -independent MET auto-phosphorylation in U87-MG glioblastoma cells and NCI-H441 lung carcinoma cells.
- Published
- 2023
3. Supplementary Table 1 from Tivantinib (ARQ197) Displays Cytotoxic Activity That Is Independent of Its Ability to Bind MET
- Author
-
Paolo Michieli, Guido Serini, Donatella Valdembri, Alberto Bardelli, Sabrina Arena, Cristina Chiriaco, Elisa Vigna, Selma Pennacchietti, and Cristina Basilico
- Abstract
PDF File - 61K, Drugs used in this study.
- Published
- 2023
4. Supplementary Figure 3 from Tivantinib (ARQ197) Displays Cytotoxic Activity That Is Independent of Its Ability to Bind MET
- Author
-
Paolo Michieli, Guido Serini, Donatella Valdembri, Alberto Bardelli, Sabrina Arena, Cristina Chiriaco, Elisa Vigna, Selma Pennacchietti, and Cristina Basilico
- Abstract
PDF File - 149K, MET exon 16 knock-out cells express a kinase-inactive MET protein.
- Published
- 2023
5. Data from Tivantinib (ARQ197) Displays Cytotoxic Activity That Is Independent of Its Ability to Bind MET
- Author
-
Paolo Michieli, Guido Serini, Donatella Valdembri, Alberto Bardelli, Sabrina Arena, Cristina Chiriaco, Elisa Vigna, Selma Pennacchietti, and Cristina Basilico
- Abstract
Purpose: MET, the high-affinity receptor for hepatocyte growth factor, is frequently deregulated in human cancer. Tivantinib (ARQ197; Arqule), a staurosporine derivative that binds to the dephosphorylated MET kinase in vitro, is being tested clinically as a highly selective MET inhibitor. However, the mechanism of action of tivantinib is still unclear.Experimental Design: The activity of tivantinib was analyzed in multiple cellular models, including: cells displaying c-MET gene amplification, strictly ‘addicted’ to MET signaling; cells with normal c-MET gene copy number, not dependent on MET for growth; cells not expressing MET; somatic knockout cells in which the ATP-binding cleft of MET, where tivantinib binds, was deleted by homologous recombination; and a cell system ‘poisoned’ by MET kinase hyperactivation, where cells die unless cultured in the presence of a specific MET inhibitor.Results: Tivantinib displayed cytotoxic activity independently of c-MET gene copy number and regardless of the presence or absence of MET. In both wild-type and isogenic knockout cells, tivantinib perturbed microtubule dynamics, induced G2/M arrest, and promoted apoptosis. Tivantinib did not rescue survival of cells ‘poisoned’ by MET kinase hyperactivation, but further incremented cell death. In all cell models analyzed, tivantinib did not inhibit HGF-dependent or -independent MET tyrosine autophosphorylation.Conclusions: We conclude that tivantinib displays cytotoxic activity via molecular mechanisms that are independent from its ability to bind MET. This notion has a relevant impact on the interpretation of clinical results, on the design of future clinical trials, and on the selection of patients receiving tivantinib treatment. Clin Cancer Res; 19(9); 2381–92. ©2013 AACR.
- Published
- 2023
6. Supplementary Information from 'Active' Cancer Immunotherapy by Anti-Met Antibody Gene Transfer
- Author
-
Paolo M. Comoglio, Selma Pennacchietti, Cristina Basilico, Lara Fontani, Cristina Chiriaco, Massimiliano Mazzone, Giovanni Pacchiana, and Elisa Vigna
- Abstract
Supplementary Information from “Active” Cancer Immunotherapy by Anti-Met Antibody Gene Transfer
- Published
- 2023
7. Supplementary Figure 1 from Microenvironment-Derived HGF Overcomes Genetically Determined Sensitivity to Anti-MET Drugs
- Author
-
Paolo Michieli, Livio Trusolino, Paolo M. Comoglio, Timothy Perera, Jeno Gyuris, May Han, William M. Rideout, Andrea Bertotti, Manuela Cazzanti, and Selma Pennacchietti
- Abstract
Supplementary Fig. 1. HGF protects MET-addicted tumor cells against MET TKI-induced growth inhibition.
- Published
- 2023
8. Supplementary Figure 3 from Microenvironment-Derived HGF Overcomes Genetically Determined Sensitivity to Anti-MET Drugs
- Author
-
Paolo Michieli, Livio Trusolino, Paolo M. Comoglio, Timothy Perera, Jeno Gyuris, May Han, William M. Rideout, Andrea Bertotti, Manuela Cazzanti, and Selma Pennacchietti
- Abstract
Supplementary Fig. 3. HGF restores physiological, GAB-1-mediated MET signaling in DN30 Fab-treated MET-addicted cells.
- Published
- 2023
9. Supplementary Figure 6 from Microenvironment-Derived HGF Overcomes Genetically Determined Sensitivity to Anti-MET Drugs
- Author
-
Paolo Michieli, Livio Trusolino, Paolo M. Comoglio, Timothy Perera, Jeno Gyuris, May Han, William M. Rideout, Andrea Bertotti, Manuela Cazzanti, and Selma Pennacchietti
- Abstract
Supplementary Fig. 6. HGF-induced activation of the PI3K pathway inversely correlates with response to MET-targeted drugs.
- Published
- 2023
10. Supplementary Methods from Microenvironment-Derived HGF Overcomes Genetically Determined Sensitivity to Anti-MET Drugs
- Author
-
Paolo Michieli, Livio Trusolino, Paolo M. Comoglio, Timothy Perera, Jeno Gyuris, May Han, William M. Rideout, Andrea Bertotti, Manuela Cazzanti, and Selma Pennacchietti
- Abstract
Supplementary Methods. Supplementary information on drugs and chemicals, human HGF knock-in mice, in vivo signal transduction analysis, study approval, and statistical analysis.
- Published
- 2023
11. Supplementary Figure 5 from Microenvironment-Derived HGF Overcomes Genetically Determined Sensitivity to Anti-MET Drugs
- Author
-
Paolo Michieli, Livio Trusolino, Paolo M. Comoglio, Timothy Perera, Jeno Gyuris, May Han, William M. Rideout, Andrea Bertotti, Manuela Cazzanti, and Selma Pennacchietti
- Abstract
Supplementary Fig. 5. Human HGF knock-in SCID mice display a physiological HGF expression pattern.
- Published
- 2023
12. Supplementary Figure 4 from Microenvironment-Derived HGF Overcomes Genetically Determined Sensitivity to Anti-MET Drugs
- Author
-
Paolo Michieli, Livio Trusolino, Paolo M. Comoglio, Timothy Perera, Jeno Gyuris, May Han, William M. Rideout, Andrea Bertotti, Manuela Cazzanti, and Selma Pennacchietti
- Abstract
Supplementary Fig. 4. Low concentrations of HGF are sufficient to fully antagonize DN30 Fab-induced EBC-1 lung carcinoma cell growth inhibition.
- Published
- 2023
13. Supplementary Figure 2 from Microenvironment-Derived HGF Overcomes Genetically Determined Sensitivity to Anti-MET Drugs
- Author
-
Paolo Michieli, Livio Trusolino, Paolo M. Comoglio, Timothy Perera, Jeno Gyuris, May Han, William M. Rideout, Andrea Bertotti, Manuela Cazzanti, and Selma Pennacchietti
- Abstract
Supplementary Fig. 2. HGF sustains HER3-independent PI3K signaling in c-MET-amplified cells exposed to MET tyrosine kinase inhibitors.
- Published
- 2023
14. Microenvironment-derived HGF overcomes genetically determined sensitivity to anti-MET drugs
- Author
-
Selma Pennacchietti, Paolo M. Comoglio, May Han, Livio Trusolino, William M. Rideout, Paolo Michieli, Jeno Gyuris, Manuela Cazzanti, Timothy Perera, and Andrea Bertotti
- Subjects
Cancer Research ,Animals ,Antibodies, Monoclonal ,Hepatocyte Growth Factor ,Mice ,Mice, SCID ,Phosphatidylinositol 3-Kinases ,Proto-Oncogene Proteins c-met ,Receptor, ErbB-3 ,Signal Transduction ,Tumor Microenvironment ,Oncology ,Medicine (all) ,Biology ,SCID ,Receptor tyrosine kinase ,Antibodies ,Ficlatuzumab ,ErbB-3 ,Monoclonal ,medicine ,PI3K/AKT/mTOR pathway ,Tumor microenvironment ,Crizotinib ,Kinase ,Cancer research ,biology.protein ,Hepatocyte growth factor ,Signal transduction ,medicine.drug ,Receptor - Abstract
Cell-based drug screenings indicate that tumors displaying c-MET gene amplification are “addicted” to MET signaling and therefore are very sensitive to MET-targeted agents. However, these screenings were conducted in the absence of the MET ligand, hepatocyte growth factor (HGF), which is abundant in the tumor microenvironment. Sensitivity of six MET-addicted human tumor cells to three MET kinase inhibitors (JNJ-38877605, PHA-665752, crizotinib) and one antagonistic anti-MET antibody (DN30 Fab) was analyzed in the absence or presence of HGF, in a stroma–tumor coculture system, and by combining anti-MET drugs with an HGF neutralizing antibody (ficlatuzumab) in human HGF knock-in mice bearing c-MET–amplified tumors. In all models examined, HGF promoted resistance to MET-targeted agents, affecting both their potency and efficacy. HGF-induced resistance was due to restoration of physiologic GAB1–mediated PI3K activation that compensated for loss of aberrant HER3-dependent PI3K signaling. Ficlatuzumab restored sensitivity to MET-targeted agents in coculture systems and overcame resistance to JNJ-38877605, crizotinib, and DN30 Fab in human HGF knock-in mice. These data suggest that c-MET–amplified tumor cells—which normally exhibit ligand-independent, constitutive MET activation—become dependent on HGF for survival upon pharmacologic MET inhibition. Because HGF is frequently overexpressed in human cancer, this mechanism may represent a major cause of resistance to anti-MET therapies. The ability of ficlatuzumab to overcome HGF-mediated resistance generates proof of principle that vertical inhibition of both a tyrosine kinase receptor and its ligand can be therapeutically beneficial and opens new perspectives for the treatment of MET-dependent tumors. Cancer Res; 74(22); 6598–609. ©2014 AACR.
- Published
- 2014
15. Tivantinib (ARQ197) Displays Cytotoxic Activity That Is Independent of Its Ability to Bind MET
- Author
-
Selma Pennacchietti, Alberto Bardelli, Donatella Valdembri, Cristina Chiriaco, Sabrina Arena, Paolo Michieli, Elisa Vigna, Cristina Basilico, and Guido Serini
- Subjects
Cancer Research ,Programmed cell death ,Oncogene ,Kinase ,medicine.medical_treatment ,Cell ,Biology ,Molecular biology ,Targeted therapy ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,oncogene ,oncology ,Cancer research ,medicine ,Cytotoxic T cell ,Hepatocyte growth factor ,Tivantinib ,medicine.drug - Abstract
Purpose: MET, the high-affinity receptor for hepatocyte growth factor, is frequently deregulated in human cancer. Tivantinib (ARQ197; Arqule), a staurosporine derivative that binds to the dephosphorylated MET kinase in vitro, is being tested clinically as a highly selective MET inhibitor. However, the mechanism of action of tivantinib is still unclear. Experimental Design: The activity of tivantinib was analyzed in multiple cellular models, including: cells displaying c-MET gene amplification, strictly ‘addicted’ to MET signaling; cells with normal c-MET gene copy number, not dependent on MET for growth; cells not expressing MET; somatic knockout cells in which the ATP-binding cleft of MET, where tivantinib binds, was deleted by homologous recombination; and a cell system ‘poisoned’ by MET kinase hyperactivation, where cells die unless cultured in the presence of a specific MET inhibitor. Results: Tivantinib displayed cytotoxic activity independently of c-MET gene copy number and regardless of the presence or absence of MET. In both wild-type and isogenic knockout cells, tivantinib perturbed microtubule dynamics, induced G2/M arrest, and promoted apoptosis. Tivantinib did not rescue survival of cells ‘poisoned’ by MET kinase hyperactivation, but further incremented cell death. In all cell models analyzed, tivantinib did not inhibit HGF-dependent or -independent MET tyrosine autophosphorylation. Conclusions: We conclude that tivantinib displays cytotoxic activity via molecular mechanisms that are independent from its ability to bind MET. This notion has a relevant impact on the interpretation of clinical results, on the design of future clinical trials, and on the selection of patients receiving tivantinib treatment. Clin Cancer Res; 19(9); 2381–92. ©2013 AACR.
- Published
- 2013
16. 'Active' cancer immunotherapy by anti-Met antibody gene transfer
- Author
-
Massimiliano Mazzone, Cristina Chiriaco, Giovanni Pacchiana, Elisa Vigna, Paolo M. Comoglio, Selma Pennacchietti, Lara Fontani, and Cristina Basilico
- Subjects
Cancer Research ,medicine.drug_class ,medicine.medical_treatment ,Genetic enhancement ,Genetic Vectors ,Monoclonal antibody ,Viral vector ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,Cell Line, Tumor ,Neoplasms ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Receptors, Growth Factor ,Phosphorylation ,030304 developmental biology ,0303 health sciences ,biology ,Lentivirus ,Gene Transfer Techniques ,Antibodies, Monoclonal ,Immunotherapy ,Genetic Therapy ,Proto-Oncogene Proteins c-met ,3. Good health ,Oncology ,030220 oncology & carcinogenesis ,Monoclonal ,biology.protein ,Cancer research ,Female ,Trans-acting ,Antibody ,Signal Transduction - Abstract
Gene therapy provides a still poorly explored opportunity to treat cancer by “active” immunotherapy as it enables the transfer of genes encoding antibodies directed against specific oncogenic proteins. By a bidirectional lentiviral vector, we transferred the cDNA encoding the heavy and light chains of a monoclonal anti-Met antibody (DN-30) to epithelial cancer cells. In vitro, the transduced cells synthesized and secreted correctly assembled antibodies with the expected high affinity, inducing down-regulation of the Met receptor and strong inhibition of the invasive growth response. The inhibitory activity resulted (a) from the interference of the antibody with the Met receptor intracellular processing (“cell autonomous activity,” in cis) and (b) from the antibody-induced cleavage of Met expressed at the cell surface (“bystander effect,” in trans). The monoclonal antibody gene transferred into live animals by systemic administration or by local intratumor delivery resulted in substantial inhibition of tumor growth. These data provide proof of concept both for targeting the Met receptor and for a gene transfer–based immunotherapy strategy. [Cancer Res 2008;68(22):9176–83]
- Published
- 2008
17. Prevention of hypoxia by myoglobin expression in human tumor cells promotes differentiation and inhibits metastasis
- Author
-
Selma Pennacchietti, Paolo Michieli, Paolo M. Comoglio, Stefania Rosano, and Maria Galluzzo
- Subjects
Cellular differentiation ,Molecular Sequence Data ,Transplantation, Heterologous ,Gene Expression ,Mice, Nude ,Biology ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Transduction, Genetic ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Neoplasm Metastasis ,030304 developmental biology ,Cell Proliferation ,DNA Primers ,0303 health sciences ,Tumor hypoxia ,Base Sequence ,Cell Death ,Neovascularization, Pathologic ,Sequence Homology, Amino Acid ,Myoglobin ,Oxygen transport ,Cell Differentiation ,General Medicine ,Tumor Oxygenation ,Hypoxia (medical) ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,Molecular biology ,Cell Hypoxia ,Recombinant Proteins ,Transplantation ,Oxygen ,030220 oncology & carcinogenesis ,Cancer cell ,Mutation ,Cancer research ,Commentary ,Female ,medicine.symptom ,Neoplasm Transplantation - Abstract
As a tumor grows, it requires increased amounts of oxygen. However, the tumor blood vessels that form to meet this demand are functionally impaired, leading to regions of hypoxia within the tumor. Such hypoxia is one of the hallmarks of malignancy and is thought to promote a number of tumorigenic properties. Here, we sought to determine how tumors without hypoxia would progress by engineering A549 human lung carcinoma cells to ectopically express myoglobin (Mb), a multifunctional heme protein that specializes in oxygen transport, storage, and buffering. Mb expression prevented the hypoxic response in vitro and delayed tumor engraftment and reduced tumor growth following xenotransplantation into mice. Experimental tumors expressing Mb displayed reduced or no hypoxia, minimal HIF-1alpha levels, and a homogeneously low vessel density. Mb-mediated tumor oxygenation promoted differentiation of cancer cells and suppressed both local and distal metastatic spreading. These effects were primarily due to reduced tumor hypoxia, because they were not observed using point-mutated forms of myoglobin unable to bind oxygen and they were abrogated by expression of a constitutively active form of HIF-1alpha. Although limited to xenograft models, these data provide experimental proof of the concept that hypoxia is not just a side effect of deregulated growth but a key factor on which the tumor relies in order to promote its own expansion.
- Published
- 2008
18. Negative feedback regulation of Met-dependent invasive growth by Notch
- Author
-
Selma Pennacchietti, Paolo M. Comoglio, M. Cristina Stella, and Livio Trusolino
- Subjects
Transcription, Genetic ,Recombinant Fusion Proteins ,Notch signaling pathway ,Down-Regulation ,Biology ,Cell Line ,Proto-Oncogene Proteins p21(ras) ,Dogs ,Downregulation and upregulation ,Cell Line, Tumor ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Drosophila Proteins ,Humans ,RNA, Messenger ,Promoter Regions, Genetic ,Molecular Biology ,Psychological repression ,Feedback, Physiological ,Homeodomain Proteins ,Transcription Factor HES-1 ,Base Sequence ,Receptors, Notch ,Hepatocyte Growth Factor ,Effector ,Membrane Proteins ,Receptor Protein-Tyrosine Kinases ,Cell Biology ,Proto-Oncogene Proteins c-met ,Cell biology ,Enzyme Activation ,Repressor Proteins ,Trachea ,Drosophila melanogaster ,Phenotype ,Notch proteins ,Hepatocyte growth factor ,Signal transduction ,Cell Division ,Signal Transduction ,medicine.drug - Abstract
The hepatocyte growth factor (HGF) receptor encoded by the Met oncogene controls a genetic program-known as "invasive growth"-responsible for several developmental processes and involved in cancer invasion and metastasis. This program functions through several regulatory gene products, as yet largely unknown, both upstream and downstream of Met. Here we show that activation of the Notch receptor results in transcriptional down-regulation of Met, suppression of HGF-dependent Ras signaling, and impairment of HGF-dependent cellular responses. In turn, Met activation leads to transcriptional induction of the Notch ligand Delta and the Notch effector HES-1, indicating that Met is able to self-tune its own protein levels and the ensuing biochemical and biological outputs through stimulation of the Notch pathway. By using branching morphogenesis of the tracheal system in Drosophila as a readout of invasive growth, we also show that exogenous expression of a constitutively active form of human Met induces enhanced sprouting of the tracheal tree, a phenotype that is further increased in embryos lacking Notch function. These results unravel an in-built mechanism of negative feedback regulation in which Met activation leads to transcriptional induction of Notch function, which in turn limits HGF activity through repression of the Met oncogene.
- Published
- 2005
19. An uncleavable form of pro-scatter factor suppresses tumor growth and dissemination in mice
- Author
-
Silvia Cavassa, Luigi Naldini, Paolo M. Comoglio, Cristina Basilico, Massimiliano Mazzone, Selma Pennacchietti, Mauro Risio, Paolo Michieli, Mazzone, M., Basilico, C., Cavassa, S., Pennacchietti, S., Risio, M., Naldini, Luigi, Comoglio, P. M., and Michieli, P.
- Subjects
Glutamine ,Cell ,lentiviral vectors ,urokinase ,Protein Engineering ,Extracellular matrix ,Mice ,gene-transfer ,Methionine ,c-met receptor ,0302 clinical medicine ,Transduction, Genetic ,Neoplasms ,serine-protease ,Neoplasm Metastasis ,Receptor ,0303 health sciences ,Hepatocyte Growth Factor ,beta-chain ,General Medicine ,Protein-Tyrosine Kinases ,Recombinant Proteins ,Extracellular Matrix ,Tumor Burden ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Hepatocyte growth factor ,Collagen ,factor/scatter factor ,invasive growth ,Tyrosine kinase ,medicine.drug ,Proteases ,purification ,Genetic Vectors ,Transplantation, Heterologous ,Mice, Nude ,Mitosis ,Breast Neoplasms ,Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,030304 developmental biology ,Carcinoma ,Lentivirus ,Genetic Therapy ,Enzyme Activation ,Transplantation ,Amino Acid Substitution ,Tumor progression ,proteolytic activation ,Cancer research ,Neoplasm Transplantation - Abstract
Scatter factor (SF), also known as hepatocyte growth factor, is ubiquitously present in the extracellular matrix of tissues in the form of an inactive precursor (pro-SF). In order to acquire biological activity pro-SF must be cleaved by specific proteases present on the cell surface. The mature form of SF controls invasive cues in both physiological and pathological processes through activation of its receptor, the Met tyrosine kinase. By substituting a single amino acid in the proteolytic site, we engineered an unprocessable form of pro-SF (uncleavable SF). Using lentivirus vector technology, we achieved local or systemic delivery of uncleavable SF in mice. We provide evidence that (a) uncleavable SF inhibits both protease-mediated pro-SF conversion and active SF-induced Met activation; (b) local expression of uncleavable SF in tumors suppresses tumor growth, impairs tumor angiogenesis, and prevents metastatic dissemination; and (c) systemic expression of uncleavable SF dramatically inhibits the growth of transplanted tumors and abolishes the formation of spontaneous metastases without perturbing vital physiological functions. These data show that proteolytic activation of pro-SF is a limiting step in tumor progression, thus suggesting a new strategy for the treatment or prevention of the malignant conversion of neoplastic lesions. ispartof: Journal of clinical investigation vol:114 issue:10 pages:1418-1432 ispartof: location:United States status: published
- Published
- 2004
20. Hepatocyte Growth Factor/Scatter Factor Receptor
- Author
-
Selma Pennacchietti and Paolo M. Comoglio
- Subjects
Hepatocyte Growth Factor Receptor ,Chemistry ,medicine ,Hepatocyte growth factor ,medicine.drug ,Cell biology - Published
- 2004
21. Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene
- Author
-
Silvia Giordano, Paolo Michieli, Selma Pennacchietti, Maria Galluzzo, Massimiliano Mazzone, and Paolo M. Comoglio
- Subjects
Cancer Research ,Transcription, Genetic ,Blotting, Western ,Molecular Sequence Data ,Fluorescent Antibody Technique ,Stimulation ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Anoxia ,Transcription (biology) ,Cell Movement ,medicine ,Tumor Cells, Cultured ,Humans ,Neoplasm Invasiveness ,RNA, Messenger ,Receptor ,Hypoxia ,Promoter Regions, Genetic ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Base Sequence ,Hepatocyte Growth Factor ,Gene Transfer Techniques ,Cell Biology ,Hypoxia (medical) ,Proto-Oncogene Proteins c-met ,Blotting, Northern ,Molecular biology ,MET PROTOONCOGENE ,3. Good health ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Hepatocyte growth factor ,medicine.symptom ,Tyrosine kinase ,medicine.drug - Abstract
Hypoxia unleashes the invasive and metastatic potential of tumor cells by largely unknown mechanisms. The Met tyrosine kinase, a high affinity receptor for hepatocyte growth factor (HGF), plays a crucial role in controlling invasive growth and is often overexpressed in cancer. Here we show that: (1) hypoxia activates transcription of the met protooncogene, resulting in higher levels of Met; (2) hypoxic areas of tumors overexpress Met; (3) hypoxia amplifies HGF signaling; (4) hypoxia synergizes with HGF in inducing invasion; (5) the proinvasive effects of hypoxia are mimicked by Met overexpression; and (6) inhibition of Met expression prevents hypoxia-induced invasive growth. These data show that hypoxia promotes tumor invasion by sensitizing cells to HGF stimulation, providing a molecular basis to explain Met overexpression in cancer. ispartof: Cancer Cell vol:3 issue:4 pages:347-361 ispartof: location:United States status: published
- Published
- 2003
22. Mutant Met-mediated transformation is ligand-dependent and can be inhibited by HGF antagonists
- Author
-
Paolo M. Comoglio, Luca Tamagnone, Antonella Maffe, Selma Pennacchietti, Silvia Giordano, Alberto Bardelli, Cristina Basilico, and Paolo Michieli
- Subjects
medicine.medical_specialty ,Cancer Research ,Recombinant Fusion Proteins ,Mutant ,Molecular Sequence Data ,Antineoplastic Agents ,Ligands ,Transfection ,Receptor tyrosine kinase ,Transformation ,Mice ,Cell surface receptor ,Internal medicine ,Proto-Oncogenes ,medicine ,Genetics ,Animals ,Humans ,Kinase activity ,Receptor ,Molecular Biology ,Tumor Stem Cell Assay ,biology ,Kinase ,Hepatocyte Growth Factor ,Tyrosine kinases ,Antibodies, Monoclonal ,Epithelial Cells ,3T3 Cells ,Oncogenes ,Proto-Oncogene Proteins c-met ,Growth factors ,Invasive growth ,Cell biology ,Endocrinology ,Cell Transformation, Neoplastic ,Amino Acid Substitution ,Drug Design ,biology.protein ,Hepatocyte growth factor ,Settore BIO/17 - ISTOLOGIA ,Tyrosine kinase ,Dimerization ,medicine.drug - Abstract
Mutations in the genes encoding for Met, Ret and Kit receptor tyrosine kinases invariably result in increased kinase activity and in the acquisition of transforming potential. However, the requirement of receptor ligands for the transformation process is still unclear. We have investigated the role of hepatocyte growth factor (HGF), the high-affinity ligand for Met, in mutant Met-mediated cell transformation. We provide evidence that the transforming potential displayed by mutant forms of Met found in human cancer is not only sensitive but entirely dependent on the presence of HGF, by showing that mutant Met transforms NIH3T3 fibroblasts, which produce endogenous HGF, but is not able to transform epithelial cells, unless exogenous HGF is supplied. Accordingly, mutant Met-induced transformation of NIH3T3 cells can be inhibited by HGF antagonists and increased by HGF stimulation. We also show that an engineered Met receptor which contains an oncogenic mutation but is impaired in its ability to bind HGF completely loses its transforming activity, which can be rescued by causing receptor dimerization using a monoclonal antibody. These results indicate that point mutations resulting in Met kinase activation are necessary but not sufficient to cause cell transformation, the latter being dependent on ligand-induced receptor dimerization. They also suggest that mutant Met-driven tumour growth depends on the availability and tissue distribution of active HGF, and provide proof-of-concept for the treatment of mutant-Met related pathologies by HGF-antagonizing drugs.
- Published
- 1999
23. Agonistic monoclonal antibodies against the Met receptor dissect the biological responses to HGF
- Author
-
Maria Prat, Tiziana Crepaldi, Selma Pennacchietti, Federico Bussolino, and Paolo M. Comoglio
- Subjects
Agonist ,medicine.drug_class ,Receptors, Cell Surface ,Ligands ,Partial agonist ,Receptor tyrosine kinase ,Cell Line ,Receptors, Urokinase Plasminogen Activator ,Mice ,Growth factor receptor ,Cell Movement ,medicine ,Enzyme-linked receptor ,Animals ,Humans ,Phosphorylation ,Insulin-like growth factor 1 receptor ,Mice, Inbred BALB C ,Binding Sites ,biology ,Hepatocyte Growth Factor ,Antibodies, Monoclonal ,Cell Biology ,Proto-Oncogene Proteins c-met ,Hepatocyte growth factor binding ,Molecular biology ,Interleukin-21 receptor ,biology.protein ,Epitopes, B-Lymphocyte ,Tyrosine - Abstract
Hepatocyte growth factor, also known as scatter factor, is a pleiotropic cytokine, which stimulates cell motility, invasion, proliferation, survival and morphogenesis, and induces the expression of specific genes by activating its receptor tyrosine kinase. In this work we have isolated, characterized and used as agonists two monoclonal antibodies (mAbs) directed against the extracellular domain of HGF receptor to investigate the requirements for receptor activation and for the different biological responses. The two mAbs display similar affinities, react with epitopes different from the hepatocyte growth factor binding site, and behave as either full or partial agonists. The full agonist mAb (DO-24) triggers all the biological effects elicited by hepatocyte growth factor, namely motility, proliferation, cell survival, invasion, tubulogenesis and angiogenesis. The partial agonist mAb (DN-30) induces only motility. Only the full agonist mAb is able to induce and sustain the expression of urokinase-type plasminogen activator receptor for prolonged periods of time, while both mAbs up-regulate the constitutive expression of urokinase-type plasminogen activator. Both mAbs activate receptor phosphorylation, which, being strictly dependent on mAb bivalence, requires receptor dimerization. Since simple receptor dimerization is not sufficient to trigger full biological responses, we propose that the region on the ss chain of the receptor recognized by the full agonist mAb is crucial for optimal receptor activation.
- Published
- 1998
24. Overexpression and activation of hepatocyte growth factor/scatter factor in human non-small-cell lung carcinomas
- Author
-
P. G. Natali, Maria Rita Nicotra, Caterina Casadio, G. Maggi, N. Arena, Maria Prat, M. F. Di Renzo, Monica Rizzo, M. Olivero, Roberto Madeddu, Paolo M. Comoglio, and Selma Pennacchietti
- Subjects
Adult ,Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Receptor expression ,Blotting, Western ,Biology ,Adenocarcinoma ,Cell surface receptor ,Carcinoma, Non-Small-Cell Lung ,Carcinoma ,medicine ,Humans ,Receptor ,Lung cancer ,Lung ,Aged ,Hepatocyte Growth Factor ,Antibodies, Monoclonal ,Receptor Protein-Tyrosine Kinases ,Middle Aged ,Proto-Oncogene Proteins c-met ,medicine.disease ,Oncology ,Cancer research ,Carcinoma, Squamous Cell ,Immunohistochemistry ,Hepatocyte growth factor ,Female ,medicine.drug ,Research Article - Abstract
Hepatocyte growth factor/scatter factor (HGF/SF) stimulates the invasive growth of epithelial cells via the c-MET oncogene-encoded receptor. In normal lung, both the receptor and the ligand are detected, and the latter is known to be a mitogenic and a motogenic factor for both cultured bronchial epithelial cells and non-small-cell carcinoma lines. Here, ligand and receptor expression was examined in 42 samples of primary human non-small-cell lung carcinoma of different histotype. Each carcinoma sample was compared with adjacent normal lung tissue. The Met/HGF receptor was found to be 2 to 10-fold increased in 25% of carcinoma samples (P = 0.0113). The ligand, HGF/SF, was found to be 10 to 100-fold overexpressed in carcinoma samples (P < 0.0001). Notably, while HGF/SF was occasionally detectable and found exclusively as a single-chain inactive precursor in normal tissues, it was constantly in the biologically-active heterodimeric form in carcinomas. Immunohistochemical staining showed homogeneous expression of both the receptor and the ligand in carcinoma samples, whereas staining was barely detectable in their normal counterparts. These data show that HGF/SF is overexpressed and consistently activated in non-small-cell lung carcinomas and may contribute to the invasive growth of lung cancer. Images Figure 1 Figure 2 Figure 3 Figure 4 Figure 5
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.