Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Appl Res Intellect Disabil ; 37(3): e13226, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38520180

RESUMEN

BACKGROUND: Paid employment represents a challenge for people with an intellectual disability and/or autism. This paper analyses the quality of jobs offered by the Engage to Change project and their relationship to the 'typicalness' of the employment offered. METHOD: Data on the quality of 384 paid jobs were collected, including hours worked and wages earned, and reported social integration at work. The typicalness of the employment experience was assessed for 141 young people, using the Index of Typicalness of Placement Questionnaire. RESULTS: The Engage to Change project offered a wide range of jobs. There is no difference in the typicalness of the employment experience in relation to the interactions in the workplace, but there are some differences in the recruitment process for men and women. CONCLUSION: Job coaches should make sure that, despite the procedures being 'atypical' for the workplace, the outcome is 'a typical employment experience' for each employee.


Asunto(s)
Empleos Subvencionados , Discapacidad Intelectual , Masculino , Humanos , Femenino , Adolescente , Lugar de Trabajo , Encuestas y Cuestionarios
2.
J Appl Res Intellect Disabil ; 37(2): e13194, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38369315

RESUMEN

BACKGROUND: The study reports the impact of the Covid-19 pandemic and lockdowns on jobs for people with intellectual disabilities and autism. The study focuses on the impact of the first and the fire-break lockdowns and the actions taken to support young people. METHOD: Data was collected from the cohort of young people currently working in Wales, and that received job coach support from the Engage to Change Project, on furlough arrangements, job retainment and job losses. Innovative initiatives to support young people are described. RESULTS: Review of the working situation during the pandemic was conducted for 184 jobs, evaluating the proportion of young people being furloughed or working remotely and compared with the general population in Wales. CONCLUSIONS: Supported employment agencies adapted their practice during the COVID-19 pandemic, offering new and innovative ways to support young people and facilitate their return to work.


Asunto(s)
Trastorno Autístico , COVID-19 , Discapacidad Intelectual , Humanos , Adolescente , COVID-19/epidemiología , Trastorno Autístico/epidemiología , Pandemias , Discapacidad Intelectual/epidemiología , Gales/epidemiología , Control de Enfermedades Transmisibles
3.
J Exp Clin Cancer Res ; 42(1): 310, 2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993874

RESUMEN

BACKGROUND: Even acknowledging the game-changing results achieved in the treatment of metastatic melanoma with the use of immune checkpoint inhibitors (ICI), a large proportion of patients (40-60%) still fail to respond or relapse due to the development of resistance. Alterations in the expression of Human Leukocyte Antigen class I (HLA-I) molecules are considered to play a major role in clinical resistance to ICI. Cellular immunotherapy with HLA-independent CAR-redirected lymphocytes is a promising alternative in this challenging setting and dedicated translational models are needed. METHODS: In this study, we propose an HLA-independent therapeutic strategy with Cytokine Induced Killer lymphocytes (CIK) genetically engineered with a Chimeric Antigen Receptor (CAR) targeting the tumor antigen CSPG4 as effector mechanism. We investigated the preclinical antitumor activity of CSPG4-CAR.CIK in vitro and in a xenograft murine model focusing on patient-derived melanoma cell lines (Mel) with defective expression of HLA-I molecules. RESULTS: We successfully generated CSPG4-CAR.CIK from patients with metastatic melanoma and reported their intense activity in vitro against a panel of CSPG4-expressing patient-derived Mel. The melanoma killing activity was intense, even at very low effector to target ratios, and not influenced by the expression level (high, low, defective) of HLA-I molecules on target cells. Furthermore, CAR.CIK conditioned medium was capable of upregulating the expression of HLA-I molecules on melanoma cells. A comparable immunomodulatory effect was replicated by treatment of Mel cells with exogenous IFN-γ and IFN-α. The antimelanoma activity of CSPG4-CAR.CIK was successfully confirmed in vivo, obtaining a significant tumor growth inhibition of an HLA-defective Mel xenograft in immunodeficient mice. CONCLUSIONS: In this study we reported the intense preclinical activity of CSPG4-CAR.CIK against melanoma, including those with low or defective HLA-I expression. Our findings support CSPG4 as a valuable CAR target in melanoma and provide translational rationale for clinical studies exploring CAR-CIK cellular immunotherapies within the challenging setting of patients not responsive or relapsing to immune checkpoint inhibitors.


Asunto(s)
Melanoma , Receptores Quiméricos de Antígenos , Humanos , Animales , Ratones , Citocinas , Receptores Quiméricos de Antígenos/genética , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia Adoptiva/métodos , Recurrencia Local de Neoplasia , Melanoma/genética , Melanoma/terapia , Inmunoterapia , Linfocitos/patología , Proteínas de la Membrana , Proteoglicanos Tipo Condroitín Sulfato
4.
Cancers (Basel) ; 15(10)2023 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-37345079

RESUMEN

Cancer cell dissemination is sustained by cell-autonomous and non-cell-autonomous functions. To disentangle the role of HGF (Hepatocyte Growth Factor) and MET ligand/receptor axis in this complex process, we genetically knocked out the MET gene in cancer cells in which MET is not the oncogenic driver. In this way, we evaluated the contribution of the HGF/MET axis to cancer cell dissemination independently of its direct activities in cells of the tumor microenvironment. The lack of MET expression in MET-/- cells has been proved by molecular characterization. From a functional point of view, HGF stimulation of MET-/- cancer cells was ineffective in eliciting intracellular signaling and in sustaining biological functions predictive of malignancy in vitro (i.e., anchorage-independent growth, invasion, and survival in the absence of matrix adhesion). Cancer cell dissemination was assessed in vivo, evaluating: (i) the ability of MET-/- lung carcinoma cells to colonize the lungs following intravenous injection and (ii) the spontaneous dissemination to distant organs of MET-/- pancreatic carcinoma cells upon orthotopic injection. In both experimental models, MET ablation affects the time of onset, the number, and the size of metastatic lesions. These results define a crucial contribution of the HGF/MET axis to cell-autonomous functions driving the metastatic process.

5.
J Appl Res Intellect Disabil ; 36(4): 787-795, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36919902

RESUMEN

BACKGROUND: People with an intellectual disability and/or autism experience low employment rates compared to the general population. This study shows what the determinants of success are in getting this group of young people into paid employment. METHOD: The research is based on data collected on 1008 young people, aged 16-25, participating in the Engage to Change project across Wales, to support young people to achieve employment. A real-time data collection system was used from their engagement and throughout their journey to employment. RESULTS: This research indicates an overall employment rate of 23% for the project to date, based on total referral. Young people who engaged in 'significant work experience', such as paid placement or supported internship, had a greater employment rate of 37%. CONCLUSION: Previous and current real work experiences increased the chances of young people of becoming employed, above the effect of supported employment and job coach support.


Asunto(s)
Trastorno Autístico , Empleos Subvencionados , Discapacidad Intelectual , Humanos , Adolescente , Gales , Demografía
6.
Clin Cancer Res ; 29(3): 621-634, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36165915

RESUMEN

PURPOSE: Antibodies against the lymphocyte PD-1 (aPD-1) receptor are cornerstone agents for advanced non-small cell lung cancer (NSCLC), based on their ability to restore the exhausted antitumor immune response. Our study reports a novel, lymphocyte-independent, therapeutic activity of aPD-1 against NSCLC, blocking the tumor-intrinsic PD-1 receptors on chemoresistant cells. EXPERIMENTAL DESIGN: PD-1 in NSCLC cells was explored in vitro at baseline, including stem-like pneumospheres, and following treatment with cisplatin both at transcriptional and protein levels. PD-1 signaling and RNA sequencing were assessed. The lymphocyte-independent antitumor activity of aPD-1 was explored in vitro, by PD-1 blockade and stimulation with soluble ligand (PD-L1s), and in vivo within NSCLC xenograft models. RESULTS: We showed the existence of PD-1+ NSCLC cell subsets in cell lines and large in silico datasets (Cancer Cell Line Encyclopedia and The Cancer Genome Atlas). Cisplatin significantly increased PD-1 expression on chemo-surviving NSCLC cells (2.5-fold P = 0.0014), while the sequential treatment with anti-PD-1 Ab impaired their recovery after chemotherapy. PD-1 was found to be associated with tumor stemness features. PD-1 expression was enhanced in NSCLC stem-like pneumospheres (P < 0.0001), significantly promoted by stimulation with soluble PD-L1 (+27% ± 4, P < 0.0001) and inhibited by PD-1 blockade (-30% ± 3, P < 0.0001). The intravenous monotherapy with anti-PD-1 significantly inhibited tumor growth of NSCLC xenografts in immunodeficient mice, without the contribution of the immune system, and delayed the occurrence of chemoresistance when combined with cisplatin. CONCLUSIONS: We report first evidence of a novel lymphocyte-independent activity of anti-PD-1 antibodies in NSCLC, capable of inhibiting chemo-surviving NSCLC cells and exploitable to contrast disease relapses following chemotherapy. See related commentary by Augustin et al., p. 505.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Ratones , Animales , Neoplasias Pulmonares/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Cisplatino/farmacología , Cisplatino/uso terapéutico , Recurrencia Local de Neoplasia , Linfocitos/metabolismo , Línea Celular Tumoral
7.
J Exp Clin Cancer Res ; 41(1): 309, 2022 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-36271379

RESUMEN

BACKGROUND: Aberrant activation of the MET receptor in cancer is sustained by genetic alterations or, more frequently, by transcriptional upregulations. A fraction of MET-amplified or mutated tumors are sensible to MET targeting agents, but their responsiveness is typically short-lasting, as secondary resistance eventually occurs. Since in the absence of genetic alterations MET is usually not a tumor driver, MET overexpressing tumors are not/poorly responsive to MET targeted therapies. Consequently, the vast majority of tumors exhibiting MET activation still represent an unmet medical need. METHODS: Here we propose an immunotherapy strategy based on T lymphocytes expressing a Chimeric Antigen Receptor (CAR) targeting MET overexpressing tumors of different histotypes. We engineered two different MET-CAR constructs and tested MET-CAR-T cell cytotoxic activity against different MET overexpressing models, including tumor cell lines, primary cancer cells, organoids, and xenografts in immune-deficient mice. RESULTS: We proved that MET-CAR-T exerted a specific cytotoxic activity against MET expressing cells. Cell killing was proportional to the level of MET expressed on the cell surface. While CAR-T cytotoxicity was minimal versus cells carrying MET at physiological levels, essentially sparing normal cells, the activity versus MET overexpressing tumors was robust, significantly controlling tumor cell growth in vitro and in vivo. Notably, MET-CAR-T cells were also able to brake acquired resistance to MET targeting agents in MET amplified cancer cells carrying secondary mutations in downstream signal transducers. CONCLUSIONS: We set and validated at the pre-clinical level a MET-CAR immunotherapy strategy potentially beneficial for cancers not eligible for MET targeted therapy with inhibitory molecules, including those exhibiting primary or secondary resistance.


Asunto(s)
Receptores Quiméricos de Antígenos , Humanos , Ratones , Animales , Inmunoterapia , Linfocitos T , Línea Celular Tumoral , Xenoinjertos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Exp Clin Cancer Res ; 41(1): 112, 2022 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-35351166

RESUMEN

BACKGROUND: The tyrosine kinase receptor encoded by the MET oncogene is a major player in cancer. When MET is responsible for the onset and progression of the transformed phenotype (MET-addicted cancers), an efficient block of its oncogenic activation results in potent tumor growth inhibition. METHODS: Here we describe a molecular engineered MET antibody (hOA-DN30) and validate its pharmacological activity in MET-addicted cancer models in vitro and in vivo. Pharmacokinetics and safety profile in non-human primates have also been assessed. RESULTS: hOA-DN30 efficiently impaired MET activation and the intracellular signalling cascade by dose and time dependent removal of the receptor from the cell surface (shedding). In vitro, the antibody suppressed cell growth by blocking cell proliferation and by concomitantly inducing cell death in multiple MET-addicted human tumor cell lines. In mice xenografts, hOA-DN30 induced an impressive reduction of tumor masses, with a wide therapeutic window. Moreover, the antibody showed high therapeutic efficacy against patient-derived xenografts generated from MET-addicted gastric tumors, leading to complete tumor regression and long-lasting effects after treatment discontinuation. Finally, hOA-DN30 showed a highly favorable pharmacokinetic profile and substantial tolerability in Cynomolgus monkeys. CONCLUSIONS: hOA-DN30 unique ability to simultaneously erase cell surface MET and release the 'decoy' receptor extracellular region results in a paramount MET blocking action. Its remarkable efficacy in a large number of pre-clinical models, as well as its pharmacological features and safety profile in non-human primates, strongly envisage a successful clinical application of this novel single-arm MET therapeutic antibody for the therapy of MET-addicted cancers.


Asunto(s)
Proteínas Proto-Oncogénicas c-met , Neoplasias Gástricas , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal
10.
Front Immunol ; 12: 775151, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925346

RESUMEN

The Hepatocyte growth factor (HGF) and its receptor (MET) promote several physiological activities such as tissue regeneration and protection from cell injury of epithelial, endothelial, neuronal and muscle cells. The therapeutic potential of MET activation has been scrutinized in the treatment of acute tissue injury, chronic inflammation, such as renal fibrosis and multiple sclerosis (MS), cardiovascular and neurodegenerative diseases. On the other hand, the HGF-MET signaling pathway may be caught by cancer cells and turned to work for invasion, metastasis, and drug resistance in the tumor microenvironment. Here, we engineered a recombinant antibody (RDO24) and two derived fragments, binding the extracellular domain (ECD) of the MET protein. The antibody binds with high affinity (8 nM) to MET ECD and does not cross-react with the closely related receptors RON nor with Semaphorin 4D. Deletion mapping studies and computational modeling show that RDO24 binds to the structure bent on the Plexin-Semaphorin-Integrin (PSI) domain, implicating the PSI domain in its binding to MET. The intact RDO24 antibody and the bivalent Fab2, but not the monovalent Fab induce MET auto-phosphorylation, mimicking the mechanism of action of HGF that activates the receptor by dimerization. Accordingly, the bivalent recombinant molecules induce HGF biological responses, such as cell migration and wound healing, behaving as MET agonists of therapeutic interest in regenerative medicine. In vivo administration of RDO24 in the murine model of MS, represented by experimental autoimmune encephalomyelitis (EAE), delays the EAE onset, mitigates the early clinical symptoms, and reduces inflammatory infiltrates. Altogether, these results suggest that engineered RDO24 antibody may be beneficial in multiple sclerosis and possibly other types of inflammatory disorders.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Desarrollo de Medicamentos , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Ingeniería de Proteínas , Proteínas Recombinantes , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/uso terapéutico , Afinidad de Anticuerpos , Antineoplásicos Inmunológicos , Línea Celular , Clonación Molecular , Desarrollo de Medicamentos/métodos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Expresión Génica , Vectores Genéticos/genética , Humanos , Ratones , Modelos Moleculares , Mutagénesis , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/genética , Relación Estructura-Actividad , Resultado del Tratamiento
11.
Cancers (Basel) ; 13(14)2021 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-34298732

RESUMEN

Pancreatic ductal adenocarcinoma is an aggressive tumor characterized by the presence of an abundant stromal compartment contributing significantly to the malignant phenotype. Pancreatic stellate cells are peculiar fibroblasts present in the stroma and represent the predominant source of extracellular matrix proteins, pro-inflammatory cytokines, and growth factors, including hepatocyte growth factor (HGF). Exploiting a co-culture system of human pancreatic stellate cells and cancer cells, we demonstrated that fibroblast activation was reduced upon HGF/MET axis inhibition. To unveil the signaling pathways sustaining stroma modulation orchestrated by MET activation in the tumor, we analyzed the gene expression profile in pancreatic cancer cells stimulated with HGF and treated with HGF/MET inhibitors. Transcriptome analysis showed that, among all the genes modulated by HGF, a subset of 125 genes was restored to the basal level following treatment with the inhibitors. By examining these genes via ingenuity pathway analysis, tenascin C emerged as a promising candidate linking MET signaling and tumor microenvironment. MET-dependent tenascin C modulation in pancreatic cancer cells was validated at RNA and protein levels both in vitro and in vivo. In conclusion, this work identifies tenascin C as a gene modulated by MET activation, suggesting a role in MET-mediated tumor-stroma interplay occurring during pancreatic tumor progression.

12.
J Exp Clin Cancer Res ; 40(1): 32, 2021 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-33446252

RESUMEN

BACKGROUND: The receptor encoded by the MET oncogene and its ligand Hepatocyte Growth Factor (HGF) are at the core of the invasive-metastatic behavior. In a number of instances genetic alterations result in ligand-independent onset of malignancy (MET addiction). More frequently, ligand stimulation of wild-type MET contributes to progression toward metastasis (MET expedience). Thus, while MET inhibitors alone are effective in the first case, combination therapy with ligand inhibitors is required in the second condition. METHODS: In this paper, we generated hybrid molecules gathering HGF and MET inhibitory properties. This has been achieved by 'head-to-tail' or 'tail-to-head' fusion of a single chain Fab derived from the DN30 MET antibody with a recombinant 'ad-hoc' engineered MET extracellular domain (decoyMET), encompassing the HGF binding site but lacking the DN30 epitope. RESULTS: The hybrid molecules correctly bind MET and HGF, inhibit HGF-induced MET downstream signaling, and quench HGF-driven biological responses, such as growth, motility and invasion, in cancer cells of different origin. Two metastatic models were generated in mice knocked-in by the human HGF gene: (i) orthotopic transplantation of pancreatic cancer cells; (ii) subcutaneous injection of primary cells derived from a cancer of unknown primary. Treatment with hybrid molecules strongly affects time of onset, number, and size of metastatic lesions. CONCLUSION: These results provide a strategy to treat metastatic dissemination driven by the HGF/MET axis.


Asunto(s)
Inmunoconjugados/farmacología , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Células A549 , Animales , Sitios de Unión de Anticuerpos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Factor de Crecimiento de Hepatocito/inmunología , Humanos , Inmunoconjugados/inmunología , Fragmentos Fab de Inmunoglobulinas/inmunología , Ratones , Ratones SCID , Metástasis de la Neoplasia , Neoplasias/inmunología , Proteínas Proto-Oncogénicas c-met/inmunología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Clin Cancer Res ; 26(23): 6321-6334, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32900797

RESUMEN

PURPOSE: No effective therapy is available for unresectable soft-tissue sarcomas (STS). This unmet clinical need prompted us to test whether chondroitin sulfate proteoglycan 4 (CSPG4)-specific chimeric antigen receptor (CAR)-redirected cytokine-induced killer lymphocytes (CAR.CIK) are effective in eliminating tumor cells derived from multiple STS histotypes in vitro and in immunodeficient mice. EXPERIMENTAL DESIGN: The experimental platform included patient-derived CAR.CIK and cell lines established from multiple STS histotypes. CAR.CIK were transduced with a retroviral vector encoding second-generation CSPG4-specific CAR (CSPG4-CAR) with 4-1BB costimulation. The functional activity of CSPG4-CAR.CIK was explored in vitro, in two- and three-dimensional STS cultures, and in three in vivo STS xenograft models. RESULTS: CSPG4-CAR.CIK were efficiently generated from patients with STS. CSPG4 was highly expressed in multiple STS histotypes by in silico analysis and on all 16 STS cell lines tested by flow cytometry. CSPG4-CAR.CIK displayed superior in vitro cytolytic activity against multiple STS histotypes as compared with paired unmodified control CIK. CSPG4-CAR.CIK also showed strong antitumor activity against STS spheroids; this effect was associated with tumor recruitment, infiltration, and matrix penetration. CSPG4-CAR.CIK significantly delayed or reversed tumor growth in vivo in three STS xenograft models (leiomyosarcoma, undifferentiated pleomorphic sarcoma, and fibrosarcoma). Tumor growth inhibition persisted for up to 2 weeks following the last administration of CSPG4-CAR.CIK. CONCLUSIONS: This study has shown that CSPG4-CAR.CIK effectively targets multiple STS histotypes in vitro and in immunodeficient mice. These results provide a strong rationale to translate the novel strategy we have developed into a clinical setting.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Células Asesinas Inducidas por Citocinas/inmunología , Inmunoterapia Adoptiva/métodos , Linfocitos/inmunología , Proteínas de la Membrana/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Sarcoma/terapia , Animales , Apoptosis , Proliferación Celular , Proteoglicanos Tipo Condroitín Sulfato/genética , Femenino , Humanos , Interleucina-2/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Sarcoma/inmunología , Sarcoma/metabolismo , Sarcoma/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cancers (Basel) ; 12(3)2020 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-32245152

RESUMEN

The MET oncogene encodes a tyrosine kinase receptor involved in the control of a complex network of biological responses that include protection from apoptosis and stimulation of cell growth during embryogenesis, tissue regeneration, and cancer progression. We previously developed an antagonist antibody (DN30) inducing the physical removal of the receptor from the cell surface and resulting in suppression of the biological responses to MET. In its bivalent form, the antibody displayed a residual agonist activity, due to dimerization of the lingering receptors, and partial activation of the downstream signaling cascade. The balance between the two opposing activities is variable in different biological systems and is hardly predictable. In this study, we generated and characterized two single-chain antibody fragments derived from DN30, sharing the same variable regions but including linkers different in length and composition. The two engineered molecules bind MET with high affinity but induce different biological responses. One behaves as a MET-antagonist, promoting programmed cell death in MET "addicted" cancer cells. The other acts as a hepatocyte growth factor (HGF)-mimetic, protecting normal cells from doxorubicin-induced apoptosis. Thus, by engineering the same receptor antibody, it is possible to generate molecules enhancing or inhibiting apoptosis either to kill cancer cells or to protect healthy tissues from the injuries of chemotherapy.

15.
Br J Cancer ; 120(5): 527-536, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30723303

RESUMEN

BACKGROUND: Interferon-induced expression of programmed cell death ligands (PD-L1/PD-L2) may sustain tumour immune-evasion. Patients featuring MET amplification, a genetic lesion driving transformation, may benefit from anti-MET treatment. We explored if MET-targeted therapy interferes with Interferon-γ modulation of PD-L1/PD-L2 in MET-amplified tumours. METHODS: PD-L1/PD-L2 expression and signalling pathways downstream of MET or Interferon-γ were analysed in MET-amplified tumour cell lines and in patient-derived tumour organoids, in basal condition, upon Interferon-γ stimulation, and after anti-MET therapy. RESULTS: PD-L1 and PD-L2 were upregulated in MET-amplified tumour cells upon Interferon-γ treatment. This induction was impaired by JNJ-605, a selective inhibitor of MET kinase activity, and MvDN30, an antibody inducing MET proteolytic cleavage. We found that activation of JAKs/ STAT1, signal transducers downstream of the Interferon-γ receptor, was neutralised by MET inhibitors. Moreover, JAK2 and MET associated in the same signalling complex depending on MET phosphorylation. Results were confirmed in MET-amplified organoids derived from human colorectal tumours, where JNJ-605 treatment revoked Interferon-γ induced PD-L1 expression. CONCLUSIONS: These data show that in MET-amplified cancers, treatment with MET inhibitors counteracts the induction of PD-1 ligands by Interferon-γ. Thus, therapeutic use of anti-MET drugs may provide additional clinical benefit over and above the intended inhibition of the target oncogene.


Asunto(s)
Antígeno B7-H1/efectos de los fármacos , Interferón gamma/farmacología , Neoplasias/genética , Proteína 2 Ligando de Muerte Celular Programada 1/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Escape del Tumor/efectos de los fármacos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Humanos , Quinasas Janus/efectos de los fármacos , Quinasas Janus/metabolismo , Neoplasias Hepáticas/secundario , Terapia Molecular Dirigida , Neoplasias/metabolismo , Organoides , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Receptores de Interferón , Factor de Transcripción STAT1/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Escape del Tumor/genética , Receptor de Interferón gamma
16.
Int J Mol Sci ; 19(12)2018 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-30544501

RESUMEN

The 'onco-receptor' MET (Hepatocyte Growth Factor Receptor) is involved in the activation of the invasive growth program that is essential during embryonic development and critical for wound healing and organ regeneration during adult life. When aberrantly activated, MET and its stroma-secreted ligand HGF (Hepatocyte Growth Factor) concur to tumor onset, progression, and metastasis in solid tumors, thus representing a relevant target for cancer precision medicine. In the vast majority of tumors, wild-type MET behaves as a 'stress-response' gene, and relies on ligand stimulation to sustain cancer cell 'scattering', invasion, and protection form apoptosis. Moreover, the MET/HGF axis is involved in the crosstalk between cancer cells and the surrounding microenvironment. Pancreatic cancer (namely, pancreatic ductal adenocarcinoma, PDAC) is an aggressive malignancy characterized by an abundant stromal compartment that is associated with early metastases and resistance to conventional and targeted therapies. Here, we discuss the role of the MET/HGF axis in tumor progression and dissemination considering as a model pancreatic cancer, and provide a proof of concept for the application of dual MET/HGF inhibition as an adjuvant therapy in pancreatic cancer patients.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Factor de Crecimiento de Hepatocito/genética , Humanos , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-met/genética
17.
Int J Cancer ; 143(7): 1774-1785, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29693242

RESUMEN

MET, a master gene sustaining "invasive growth," is a relevant target for cancer precision therapy. In the vast majority of tumors, wild-type MET behaves as a "stress-response" gene and relies on the ligand (HGF) to sustain cell "scattering," invasive growth and apoptosis protection (oncogene "expedience"). In this context, concomitant targeting of MET and HGF could be crucial to reach effective inhibition. To test this hypothesis, we combined an anti-MET antibody (MvDN30) inducing "shedding" (i.e., removal of MET from the cell surface), with a "decoy" (i.e., the soluble extracellular domain of the MET receptor) endowed with HGF-sequestering ability. To avoid antibody/decoy interaction-and subsequent neutralization-we identified a single aminoacid in the extracellular domain of MET-lysine 842-that is critical for MvDN30 binding and engineered the corresponding recombinant decoyMET (K842E). DecoyMETK842E retains the ability to bind HGF with high affinity and inhibits HGF-induced MET phosphorylation. In HGF-dependent cellular models, MvDN30 antibody and decoyMETK842E used in combination cooperate in restraining invasive growth, and synergize in blocking cancer cell "scattering." The antibody and the decoy unbridle apoptosis of colon cancer stem cells grown in vitro as spheroids. In a preclinical model, built by orthotopic transplantation of a human pancreatic carcinoma in SCID mice engineered to express human HGF, concomitant treatment with antibody and decoy significantly reduces metastatic spread. The data reported indicate that vertical targeting of the MET/HGF axis results in powerful inhibition of ligand-dependent MET activation, providing proof of concept in favor of combined target therapy of MET "expedience."


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias del Colon/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Animales , Apoptosis , Proliferación Celular , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Femenino , Glioblastoma/inmunología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ligandos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Proto-Oncogénicas c-met/inmunología , Proteínas Proto-Oncogénicas c-met/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cancer Res ; 76(17): 5019-29, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27364553

RESUMEN

MET oncogene amplification is emerging as a major mechanism of acquired resistance to EGFR-directed therapy in lung and colorectal cancers. Furthermore, MET amplification predicts responsiveness to MET inhibitors currently in clinical trials. Among the anti-MET drugs available, ATP-competitive small-molecule kinase inhibitors abrogate receptor autophosphorylation and downstream activation of ERK1/2 and AKT, resulting in cell-cycle arrest. However, this antiproliferative effect allows persistence of a pool of cancer cells that are quiescent but alive. Once the inhibition is removed, rebound activation of MET-driven cell proliferative pathways and tumor growth may occur, an adverse event observed frequently in clinical settings after drug discontinuation. Here we show that inhibitor withdrawal prompts receptor phosphorylation to levels higher than those displayed at steady-state and generates a rebound effect pushing quiescent cancer cells back into the cell cycle, both in vitro and in experimental tumor models in vivo Mechanistically, we found that inhibitor treatment blocks MET endocytosis, causing a local increase in the number of receptors at the plasma membrane. Upon inhibitor washout, the receptor is readily rephosphorylated. The initial phosphorylation is not only increased but also prolonged in duration due to downmodulation of a phosphatase-mediated MET-negative feedback loop, which accompanies receptor internalization. Notably, treatment with a MET therapeutic antibody that induces proteolytic cleavage of the receptor at the cell surface substantially prevents this rebound effect, providing a rationale to combine or alternate these mechanistically different types of MET-targeted therapy. Cancer Res; 76(17); 5019-29. ©2016 AACR.


Asunto(s)
Anticuerpos/farmacología , Antineoplásicos/farmacología , Recurrencia Local de Neoplasia/patología , Neoplasias Experimentales/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Microscopía Confocal , Recurrencia Local de Neoplasia/metabolismo , Neoplasias Experimentales/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mol Oncol ; 10(6): 938-48, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27103110

RESUMEN

The kinase receptor encoded by the Met oncogene is a sensible target for cancer therapy. The chimeric monovalent Fab fragment of the DN30 monoclonal antibody (MvDN30) has an odd mechanism of action, based on cell surface removal of Met via activation of specific plasma membrane proteases. However, the short half-life of the Fab, due to its low molecular weight, is a severe limitation for the deployment in therapy. This issue was addressed by increasing the Fab molecular weight above the glomerular filtration threshold through the duplication of the constant domains, in tandem (DCD-1) or reciprocally swapped (DCD-2). The two newly engineered molecules showed biochemical properties comparable to the original MvDN30 in vitro, acting as full Met antagonists, impairing Met phosphorylation and activation of downstream signaling pathways. As a consequence, Met-mediated biological responses were inhibited, including anchorage-dependent and -independent cell growth. In vivo DCD-1 and DCD-2 showed a pharmacokinetic profile significantly improved over the original MvDN30, doubling the circulating half-life and reducing the clearance. In pre-clinical models of cancer, generated by injection of tumor cells or implant of patient-derived samples, systemic administration of the engineered molecules inhibited the growth of Met-addicted tumors.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Colon/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Fragmentos Fab de Inmunoglobulinas/uso terapéutico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Células A549 , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Antineoplásicos/sangre , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Colon/metabolismo , Colon/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Femenino , Células HEK293 , Semivida , Humanos , Fragmentos Fab de Inmunoglobulinas/sangre , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/farmacología , Ratones Endogámicos NOD , Ratones SCID , Fosforilación/efectos de los fármacos , Dominios Proteicos , Ingeniería de Proteínas , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/efectos de los fármacos
20.
J Mol Cell Cardiol ; 93: 84-97, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26924269

RESUMEN

Cardiac hypertrophy is a major risk factor for heart failure. Hence, its attenuation represents an important clinical goal. Erk1,2 signalling is pivotal in the cardiac response to stress, suggesting that its inhibition may be a good strategy to revert heart hypertrophy. In this work, we unveiled the events associated with cardiac hypertrophy by means of a transgenic model expressing activated Met receptor. c-Met proto-oncogene encodes for the tyrosine kinase receptor of Hepatocyte growth factor and is a strong inducer of Ras-Raf-Mek-Erk1,2 pathway. We showed that three weeks after the induction of activated Met, the heart presents a remarkable concentric hypertrophy, with no signs of congestive failure and preserved contractility. Cardiac enlargement is accompanied by upregulation of growth-regulating transcription factors, natriuretic peptides, cytoskeletal proteins, and Extracellular Matrix remodelling factors (Timp1 and Pai1). At a later stage, cardiac hypertrophic remodelling results into heart failure with preserved systolic function. Prevention trial by suppressing activated Met showed that cardiac hypertrophy is reversible, and progression to heart failure is prevented. Notably, treatment with Pimasertib, Mek1 inhibitor, attenuates cardiac hypertrophy and remodelling. Our results suggest that modulation of Erk1.2 signalling may constitute a new therapeutic approach for treating cardiac hypertrophies.


Asunto(s)
Cardiomegalia/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Niacinamida/análogos & derivados , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Cardiomegalia/diagnóstico , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/genética , Línea Celular , Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Uniones Comunicantes/metabolismo , Regulación de la Expresión Génica , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ratones , Ratones Transgénicos , Niacinamida/farmacología , Fenotipo , Proteínas Proto-Oncogénicas c-met/genética , Remodelación Ventricular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA