Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters











Publication year range
1.
Article in English | MEDLINE | ID: mdl-39207511

ABSTRACT

PURPOSE: Suicide is a major public health problem across the world. Extensive research on the field shows that suicide is affected by several sociological, economic, and cultural risk factors. Over the last century, social changes have driven the reshaping of traditional gender roles, often in an uneven fashion, strongly depending on context. This study proposes updated findings on the impact that changes in traditional gender roles could have on suicide rates METHODS: It will do so by examining the correlation between female labor force participation (FLPR) and sex-specific suicide rates. Moreover, it will examine this association depending on human development (HDI) and Hofstede's individualism index. To do so, data from 2010 to 2019 from 47 countries is collected from the WHO, ILOSTAT and UN agencies' websites. RESULTS: Analysis show a significant interaction between FLPR, HDI and individualism index scores on male suicide rates (p = 0.002). There is a negative association between FLPR and male suicide rates in relatively lower HDI countries, while in very high HDI countries an increase in FLPR is correlated with an increase in male suicide rates. Similar trends but no significant interaction is observed for female suicide rates. CONCLUSION: This study suggests that female participation is beneficial for male population as it reduces male suicide rates. However, this association appears to be context dependent. In countries where institutional adjustment is already established, and human development is very high, other factors might be of interest in examining the trends of suicide rates among men and women.

2.
Mol Ther ; 32(8): 2741-2761, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38894542

ABSTRACT

HER2 amplification occurs in approximately 5% of colorectal cancer (CRC) cases and is associated only partially with clinical response to combined human epidermal growth factor receptor 2 (HER2)/epidermal growth factor receptor (EGFR)-targeted treatment. An alternative approach based on adoptive cell therapy using T cells engineered with anti-HER2 chimeric antigen receptor (CAR) proved to be toxic due to on-target/off-tumor activity. Here we describe a combinatorial strategy to safely target HER2 amplification and carcinoembryonic antigen (CEA) expression in CRC using a synNotch-CAR-based artificial regulatory network. The natural killer (NK) cell line NK-92 was engineered with an anti-HER2 synNotch receptor driving the expression of a CAR against CEA only when engaged. After being transduced and sorted for HER2-driven CAR expression, cells were cloned. The clone with optimal performances in terms of specificity and amplitude of CAR induction demonstrated significant activity in vitro and in vivo specifically against HER2-amplified (HER2amp)/CEA+ CRC models, with no effects on cells with physiological HER2 levels. The HER2-synNotch/CEA-CAR-NK system provides an innovative, scalable, and safe off-the-shelf cell therapy approach with potential against HER2amp CRC resistant or partially responsive to HER2/EGFR blockade.


Subject(s)
Colorectal Neoplasms , Receptor, ErbB-2 , Receptors, Chimeric Antigen , Xenograft Model Antitumor Assays , Colorectal Neoplasms/therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Humans , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Animals , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Mice , Cell Line, Tumor , Carcinoembryonic Antigen/immunology , Carcinoembryonic Antigen/genetics , Gene Amplification , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Immunotherapy/methods , Immunotherapy, Adoptive/methods , Disease Models, Animal , Female
3.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38892318

ABSTRACT

The MET receptor is one of the main drivers of 'invasive growth', a multifaceted biological response essential during embryonic development and tissue repair that is usurped by cancer cells to induce and sustain the malignant phenotype. MET stands out as one of the most important oncogenes activated in cancer and its inhibition has been explored since the initial era of cancer-targeted therapy. Different approaches have been developed to hamper MET signaling and/or reduce MET (over)expression as a hallmark of transformation. Considering the great interest gained by cancer immunotherapy, this review evaluates the opportunity of targeting MET within therapeutic approaches based on the exploitation of immune functions, either in those cases where MET impairment is crucial to induce an effective response (i.e., when MET is the driver of the malignancy), or when blocking MET represents a way for potentiating the treatment (i.e., when MET is an adjuvant of tumor fitness).


Subject(s)
Immunotherapy , Neoplasms , Proto-Oncogene Proteins c-met , Humans , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Neoplasms/therapy , Neoplasms/genetics , Neoplasms/immunology , Immunotherapy/methods , Animals , Oncogenes , Molecular Targeted Therapy/methods , Signal Transduction
4.
J Appl Res Intellect Disabil ; 37(3): e13226, 2024 May.
Article in English | MEDLINE | ID: mdl-38520180

ABSTRACT

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.


Subject(s)
Employment, Supported , Intellectual Disability , Male , Humans , Female , Adolescent , Workplace , Surveys and Questionnaires
5.
J Appl Res Intellect Disabil ; 37(2): e13194, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38369315

ABSTRACT

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.


Subject(s)
Autistic Disorder , COVID-19 , Intellectual Disability , Humans , Adolescent , COVID-19/epidemiology , Autistic Disorder/epidemiology , Pandemics , Intellectual Disability/epidemiology , Wales/epidemiology , Communicable Disease Control
6.
J Exp Clin Cancer Res ; 42(1): 310, 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37993874

ABSTRACT

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.


Subject(s)
Melanoma , Receptors, Chimeric Antigen , Humans , Animals , Mice , Cytokines , Receptors, Chimeric Antigen/genetics , Immune Checkpoint Inhibitors , Immunotherapy, Adoptive/methods , Neoplasm Recurrence, Local , Melanoma/genetics , Melanoma/therapy , Immunotherapy , Lymphocytes/pathology , Membrane Proteins , Chondroitin Sulfate Proteoglycans
7.
Cancers (Basel) ; 15(10)2023 May 13.
Article in English | MEDLINE | ID: mdl-37345079

ABSTRACT

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.

8.
J Appl Res Intellect Disabil ; 36(4): 787-795, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36919902

ABSTRACT

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.


Subject(s)
Autistic Disorder , Employment, Supported , Intellectual Disability , Humans , Adolescent , Wales , Demography
9.
Clin Cancer Res ; 29(3): 621-634, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36165915

ABSTRACT

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.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Mice , Animals , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/pharmacology , Cisplatin/therapeutic use , Neoplasm Recurrence, Local , Lymphocytes/metabolism , Cell Line, Tumor
10.
J Exp Clin Cancer Res ; 41(1): 309, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36271379

ABSTRACT

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.


Subject(s)
Receptors, Chimeric Antigen , Humans , Mice , Animals , Immunotherapy , T-Lymphocytes , Cell Line, Tumor , Heterografts , Xenograft Model Antitumor Assays
12.
J Exp Clin Cancer Res ; 41(1): 112, 2022 Mar 29.
Article in English | MEDLINE | ID: mdl-35351166

ABSTRACT

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.


Subject(s)
Proto-Oncogene Proteins c-met , Stomach Neoplasms , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction
13.
Front Immunol ; 12: 775151, 2021.
Article in English | MEDLINE | ID: mdl-34925346

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/pharmacology , Drug Development , Hepatocyte Growth Factor/antagonists & inhibitors , Protein Engineering , Recombinant Proteins , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antibody Affinity , Antineoplastic Agents, Immunological , Cell Line , Cloning, Molecular , Drug Development/methods , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression , Genetic Vectors/genetics , Humans , Mice , Models, Molecular , Mutagenesis , Protein Engineering/methods , Recombinant Proteins/genetics , Structure-Activity Relationship , Treatment Outcome
14.
Cancers (Basel) ; 13(14)2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34298732

ABSTRACT

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.

15.
J Exp Clin Cancer Res ; 40(1): 32, 2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33446252

ABSTRACT

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.


Subject(s)
Immunoconjugates/pharmacology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , A549 Cells , Animals , Binding Sites, Antibody , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/immunology , Humans , Immunoconjugates/immunology , Immunoglobulin Fab Fragments/immunology , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasms/immunology , Proto-Oncogene Proteins c-met/immunology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Xenograft Model Antitumor Assays
16.
Clin Cancer Res ; 26(23): 6321-6334, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32900797

ABSTRACT

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.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Cytokine-Induced Killer Cells/immunology , Immunotherapy, Adoptive/methods , Lymphocytes/immunology , Membrane Proteins/metabolism , Receptors, Chimeric Antigen/immunology , Sarcoma/therapy , Animals , Apoptosis , Cell Proliferation , Chondroitin Sulfate Proteoglycans/genetics , Female , Humans , Interleukin-2/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Sarcoma/immunology , Sarcoma/metabolism , Sarcoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Cancers (Basel) ; 12(3)2020 Mar 21.
Article in English | MEDLINE | ID: mdl-32245152

ABSTRACT

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.

18.
Br J Cancer ; 120(5): 527-536, 2019 03.
Article in English | MEDLINE | ID: mdl-30723303

ABSTRACT

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.


Subject(s)
B7-H1 Antigen/drug effects , Interferon-gamma/pharmacology , Neoplasms/genetics , Programmed Cell Death 1 Ligand 2 Protein/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Tumor Escape/drug effects , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Janus Kinases/drug effects , Janus Kinases/metabolism , Liver Neoplasms/secondary , Molecular Targeted Therapy , Neoplasms/metabolism , Organoids , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Proto-Oncogene Proteins c-met/genetics , Receptors, Interferon , STAT1 Transcription Factor/drug effects , STAT1 Transcription Factor/metabolism , Signal Transduction , Tumor Escape/genetics , Interferon gamma Receptor
19.
Int J Mol Sci ; 19(12)2018 Dec 07.
Article in English | MEDLINE | ID: mdl-30544501

ABSTRACT

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.


Subject(s)
Hepatocyte Growth Factor/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , Animals , Hepatocyte Growth Factor/genetics , Humans , Neoplasm Metastasis , Proto-Oncogene Proteins c-met/genetics
20.
Int J Cancer ; 143(7): 1774-1785, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29693242

ABSTRACT

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."


Subject(s)
Antibodies, Monoclonal/pharmacology , Colonic Neoplasms/drug therapy , Glioblastoma/drug therapy , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Apoptosis , Cell Proliferation , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Ligands , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Proto-Oncogene Proteins c-met/immunology , Proto-Oncogene Proteins c-met/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL