Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 133(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38099496

RESUMEN

Cell therapies such as tumor-infiltrating lymphocyte (TIL) therapy have shown promise in the treatment of patients with refractory solid tumors, with improvement in response rates and durability of responses nevertheless sought. To identify targets capable of enhancing the antitumor activity of T cell therapies, large-scale in vitro and in vivo clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 screens were performed, with the SOCS1 gene identified as a top T cell-enhancing target. In murine CD8+ T cell-therapy models, SOCS1 served as a critical checkpoint in restraining the accumulation of central memory T cells in lymphoid organs as well as intermediate (Texint) and effector (Texeff) exhausted T cell subsets derived from progenitor exhausted T cells (Texprog) in tumors. A comprehensive CRISPR tiling screen of the SOCS1-coding region identified sgRNAs targeting the SH2 domain of SOCS1 as the most potent, with an sgRNA with minimal off-target cut sites used to manufacture KSQ-001, an engineered TIL therapy with SOCS1 inactivated by CRISPR/Cas9. KSQ-001 possessed increased responsiveness to cytokine signals and enhanced in vivo antitumor function in mouse models. These data demonstrate the use of CRISPR/Cas9 screens in the rational design of T cell therapies.


Asunto(s)
Sistemas CRISPR-Cas , Neoplasias , Humanos , Animales , Ratones , ARN Guía de Sistemas CRISPR-Cas , Linfocitos Infiltrantes de Tumor , Inmunoterapia Adoptiva , Neoplasias/genética , Edición Génica , Proteína 1 Supresora de la Señalización de Citocinas/genética
2.
J Occup Environ Med ; 64(12): 1046-1052, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-35902352

RESUMEN

OBJECTIVE: The aim of the study is to determine the associations of workers' compensation claim costs and return to work with drugs prescribed for early symptom management. METHODS: Claims filed from 1998 to 2007 were followed for 10 years from the injury date. Drugs analyzed included gabapentin, pregabalin, antipsychotics, antidepressants, sedatives, benzodiazepines, carisoprodol, and opioids, controlling for initial reserve, sex, age, physical therapy, attorney involvement, and surgery. RESULTS: Gabapentin, antipsychotics, antidepressants, and sedatives used in the first 3 months after injury were significantly associated with higher claim cost (≥$100,000). All opioid morphine equivalent doses greater than or equal to 5 mg/d for the first 6 months was significantly associated with higher cost (≥$100,000) and not being released to work at end of third year after injury with dose-response relationships. CONCLUSIONS: Prescription patterns in the first 3 months or first 6 months of workers' compensation claim development may be used as predictors of claim outcomes.


Asunto(s)
Prescripciones de Medicamentos , Humanos
3.
J Clin Invest ; 132(13)2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35552271

RESUMEN

The inability of CD8+ effector T cells (Teffs) to reach tumor cells is an important aspect of tumor resistance to cancer immunotherapy. The recruitment of these cells to the tumor microenvironment (TME) is regulated by integrins, a family of adhesion molecules that are expressed on T cells. Here, we show that 7HP349, a small-molecule activator of lymphocyte function-associated antigen-1 (LFA-1) and very late activation antigen-4 (VLA-4) integrin cell-adhesion receptors, facilitated the preferential localization of tumor-specific T cells to the tumor and improved antitumor response. 7HP349 monotherapy had modest effects on anti-programmed death 1-resistant (anti-PD-1-resistant) tumors, whereas combinatorial treatment with anti-cytotoxic T lymphocyte-associated protein 4 (anti-CTLA-4) increased CD8+ Teff intratumoral sequestration and synergized in cooperation with neutrophils in inducing cancer regression. 7HP349 intratumoral CD8+ Teff enrichment activity depended on CXCL12. We analyzed gene expression profiles using RNA from baseline and on treatment tumor samples of 14 melanoma patients. We identified baseline CXCL12 gene expression as possibly improving the likelihood or response to anti-CTLA-4 therapies. Our results provide a proof-of-principle demonstration that LFA-1 activation could convert a T cell-exclusionary TME to a T cell-enriched TME through mechanisms involving cooperation with innate immune cells.


Asunto(s)
Antígeno-1 Asociado a Función de Linfocito , Melanoma , Linfocitos T CD8-positivos , Antígeno CTLA-4 , Humanos , Inmunoterapia/métodos , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Linfocitos Infiltrantes de Tumor , Melanoma/tratamiento farmacológico , Melanoma/genética , Receptor de Muerte Celular Programada 1 , Linfocitos T/metabolismo , Microambiente Tumoral
4.
Cancer Immunol Res ; 10(4): 420-436, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35181787

RESUMEN

Protein arginine methyltransferases (PRMT) are a widely expressed class of enzymes responsible for catalyzing arginine methylation on numerous protein substrates. Among them, type I PRMTs are responsible for generating asymmetric dimethylarginine. By controlling multiple basic cellular processes, such as DNA damage responses, transcriptional regulation, and mRNA splicing, type I PRMTs contribute to cancer initiation and progression. A type I PRMT inhibitor, GSK3368715, has been developed and has entered clinical trials for solid and hematologic malignancies. Although type I PRMTs have been reported to play roles in modulating immune cell function, the immunologic role of tumor-intrinsic pathways controlled by type I PRMTs remains uncharacterized. Here, our The Cancer Genome Atlas dataset analysis revealed that expression of type I PRMTs associated with poor clinical response and decreased immune infiltration in patients with melanoma. In cancer cell lines, inhibition of type I PRMTs induced an IFN gene signature, amplified responses to IFN and innate immune signaling, and decreased expression of the immunosuppressive cytokine VEGF. In immunocompetent mouse tumor models, including a model of T-cell exclusion that represents a common mechanism of anti-programmed cell death protein 1 (PD-1) resistance in humans, type I PRMT inhibition increased T-cell infiltration, produced durable responses dependent on CD8+ T cells, and enhanced efficacy of anti-PD-1 therapy. These data indicate that type I PRMT inhibition exhibits immunomodulatory properties and synergizes with immune checkpoint blockade (ICB) to induce durable antitumor responses in a T cell-dependent manner, suggesting that type I PRMT inhibition can potentiate an antitumor immunity in refractory settings.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Proteína-Arginina N-Metiltransferasas , Animales , Arginina , Humanos , Inmunidad , Ratones , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo
6.
J Occup Environ Med ; 63(10): e694-e700, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34354021

RESUMEN

OBJECTIVE: To determine long term (11 year) trends in gabapentin and pregabalin prescribing among workers' compensation claimants at various opioid dose combinations (low, medium, high, and very high) in Louisiana. METHOD: A longitudinal study of 18,737 claimants who filled any prescriptions between 2008 and 2018. RESULTS: The proportion of claimants prescribed opioids alone at all dose levels decreased dramatically. The proportion claimants prescribed the combination of low dose opioids and low dose gabapentinoids increased (7.7% to 10.9%). Prescribing higher daily doses of gabapentinoids was associated with higher daily doses of opioids. Gabapentinoid prescribing was associated with continued prescribing of medium and high dose opioids as claims matured. CONCLUSIONS: Overall opioid prescribing decreased over time, while prescribing low dose opioids with gabapentinoids, increased.


Asunto(s)
Analgésicos Opioides , Pautas de la Práctica en Medicina , Gabapentina , Humanos , Estudios Longitudinales , Indemnización para Trabajadores
7.
J Immunother Cancer ; 9(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33589527

RESUMEN

BACKGROUND: Despite approval of immunotherapy for a wide range of cancers, the majority of patients fail to respond to immunotherapy or relapse following initial response. These failures may be attributed to immunosuppressive mechanisms co-opted by tumor cells. However, it is challenging to use conventional methods to systematically evaluate the potential of tumor intrinsic factors to act as immune regulators in patients with cancer. METHODS: To identify immunosuppressive mechanisms in non-responders to cancer immunotherapy in an unbiased manner, we performed genome-wide CRISPR immune screens and integrated our results with multi-omics clinical data to evaluate the role of tumor intrinsic factors in regulating two rate-limiting steps of cancer immunotherapy, namely, T cell tumor infiltration and T cell-mediated tumor killing. RESULTS: Our studies revealed two distinct types of immune resistance regulators and demonstrated their potential as therapeutic targets to improve the efficacy of immunotherapy. Among them, PRMT1 and RIPK1 were identified as a dual immune resistance regulator and a cytotoxicity resistance regulator, respectively. Although the magnitude varied between different types of immunotherapy, genetically targeting PRMT1 and RIPK1 sensitized tumors to T-cell killing and anti-PD-1/OX40 treatment. Interestingly, a RIPK1-specific inhibitor enhanced the antitumor activity of T cell-based and anti-OX40 therapy, despite limited impact on T cell tumor infiltration. CONCLUSIONS: Collectively, the data provide a rich resource of novel targets for rational immuno-oncology combinations.


Asunto(s)
Sistemas CRISPR-Cas , Genómica , Neoplasias/genética , Escape del Tumor/genética , Microambiente Tumoral/genética , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica/genética , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/inmunología , Neoplasias/terapia , Proteína-Arginina N-Metiltransferasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteínas Represoras/genética , Linfocitos T/inmunología , Microambiente Tumoral/inmunología
8.
J Occup Environ Med ; 63(5): 374-380, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33395171

RESUMEN

OBJECTIVE: Determine the industries with the highest proportion of accepted COVID-19 related workers' compensation (WC) claims. METHODS: Study included 21,336 WC claims (1898 COVID-19 and 19,438 other claims) that were filed between January 1, 2020 and August 31, 2020 from 11 states in the Midwest United States. RESULT: The overwhelming proportion of all COVID-19 related WC claims submitted and accepted were from healthcare workers (83.77%). Healthcare was the only industrial classification that was at significantly higher COVID-19 WC claim submission risk (odds ratio [OR]: 4.00; 95% confidence intervals [CI]: 2.77 to 5.79) controlling for type of employment, sex, age, and presumption of COVID-19 work-relatedness. Within healthcare employment, WC claims submitted by workers in medical laboratories had the highest risk (crude rate ratio of 8.78). CONCLUSION: Healthcare employment is associated with an increased risk of developing COVID-19 infections and submitting a workers' compensation claim.


Asunto(s)
COVID-19/economía , Personal de Salud/clasificación , Industrias/clasificación , Enfermedades Profesionales/economía , Indemnización para Trabajadores/estadística & datos numéricos , Adulto , Anciano , Femenino , Personal de Salud/estadística & datos numéricos , Humanos , Industrias/estadística & datos numéricos , Masculino , Personal de Laboratorio Clínico/estadística & datos numéricos , Persona de Mediana Edad , Medio Oeste de Estados Unidos/epidemiología , Oportunidad Relativa , SARS-CoV-2
9.
Cancer Immunol Immunother ; 70(4): 1101-1113, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33123754

RESUMEN

Although immunotherapy has achieved impressive durable clinical responses, many cancers respond only temporarily or not at all to immunotherapy. To find novel, targetable mechanisms of resistance to immunotherapy, patient-derived melanoma cell lines were transduced with 576 open reading frames, or exposed to arrayed libraries of 850 bioactive compounds, prior to co-culture with autologous tumor-infiltrating lymphocytes (TILs). The synergy between the targets and TILs to induce apoptosis, and the mechanisms of inhibiting resistance to TILs were interrogated. Gene expression analyses were performed on tumor samples from patients undergoing immunotherapy for metastatic melanoma. Finally, the effect of inhibiting the top targets on the efficacy of immunotherapy was investigated in multiple preclinical models. Aurora kinase was identified as a mediator of melanoma cell resistance to T-cell-mediated cytotoxicity in both complementary screens. Aurora kinase inhibitors were validated to synergize with T-cell-mediated cytotoxicity in vitro. The Aurora kinase inhibition-mediated sensitivity to T-cell cytotoxicity was shown to be partially driven by p21-mediated induction of cellular senescence. The expression levels of Aurora kinase and related proteins were inversely correlated with immune infiltration, response to immunotherapy and survival in melanoma patients. Aurora kinase inhibition showed variable responses in combination with immunotherapy in vivo, suggesting its activity is modified by other factors in the tumor microenvironment. These data suggest that Aurora kinase inhibition enhances T-cell cytotoxicity in vitro and can potentiate antitumor immunity in vivo in some but not all settings. Further studies are required to determine the mechanism of primary resistance to this therapeutic intervention.


Asunto(s)
Aurora Quinasa A/metabolismo , Aurora Quinasa B/metabolismo , Resistencia a Antineoplásicos/inmunología , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Linfocitos T Citotóxicos/trasplante , Animales , Apoptosis , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/genética , Aurora Quinasa B/antagonistas & inhibidores , Aurora Quinasa B/genética , Proliferación Celular , Femenino , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/terapia , Ratones , Pronóstico , Tasa de Supervivencia , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Cancer Ther ; 20(3): 500-511, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33361272

RESUMEN

Immune-checkpoint inhibitors and adoptive tumor-infiltrating lymphocyte (TIL) therapies have profoundly improved the survival of patients with melanoma. However, a majority of patients do not respond to these agents, and many responders experience disease relapse. Although numerous innovative treatments are being explored to offset the limitations of these agents, novel therapeutic combinations with immunotherapies have the potential to improve patient responses. In this study, we evaluated the antimelanoma activity of immunotherapy combinations with Telaglenastat (CB-839), a potent glutaminase inhibitor (GLSi) that has favorable systemic tolerance. In in vitro TIL:tumor coculture studies, CB-839 treatment improved the cytotoxic activity of autologous TILs on patient-derived melanoma cells. CB-839 treatment decreased the conversion of glutamine to alpha-ketoglutarate (αKGA) more potently in tumor cells versus TILs in these cocultures. These results suggest that CB-839 may improve immune function in a tumor microenvironment by differentially altering tumor and immune cell metabolism. In vivo CB-839 treatment activated melanoma antigen-specific T cells and improved their tumor killing activity in an immune-competent mouse model of adoptive T-cell therapy. Additionally, the combination of CB-839 with anti-PD1 or anti-CTLA4 antibodies increased tumor infiltration by effector T cells and improved the antitumor activity of these checkpoint inhibitors in a high mutation burden mouse melanoma model. Responsiveness to these treatments was also accompanied by an increase of interferon gamma (IFNγ)-associated gene expression in the tumors. Together, these results provide a strong rationale for combining CB-839 with immune therapies to improve efficacy of these treatments against melanoma.


Asunto(s)
Glutaminasa/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Melanoma/tratamiento farmacológico , Linfocitos T/metabolismo , Animales , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Ratones , Microambiente Tumoral
11.
J Occup Environ Med ; 63(2): e46-e52, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33196519

RESUMEN

OBJECTIVE: To characterize changes in opioid, gabapentin, and pregabalin utilization patterns and cost trends between 2008 and 2018 in a Louisiana workers' compensation claims population and explore the role of gabapentinoids as alternative analgesics during the opioid epidemic. METHOD: Filled prescriptions for gabapentinoids and opioids were studied for 11 years in a cohort of 18,737 claimants. RESULTS: The proportion of claimants prescribed gabapentin increased 2-fold (8.9% to 18.9%) and average drug cost per claimant decreased 22% ($612 to $480). The proportion of claimants prescribed pregabalin decreased approximately 80% (11.7% to 2.5%) and average drug cost per claim increased 224% ($911 to $2952). Proportion of claimants prescribed opioids decreased 20% (80% to 64.2%) and average drug cost per claim decreased 46% ($691 to $371). CONCLUSIONS: Utilization increased substantially for gabapentin and decreased for pregabalin and opioids.


Asunto(s)
Analgésicos Opioides , Indemnización para Trabajadores , Analgésicos/uso terapéutico , Analgésicos Opioides/uso terapéutico , Gabapentina/uso terapéutico , Humanos , Pregabalina/uso terapéutico
12.
Front Microbiol ; 11: 975, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32508787

RESUMEN

Indwelling silicone valves called voice prostheses (VPs) are the gold standard for speech rehabilitation in patients with laryngeal cancer following total laryngectomy. Reported VP lifespans amongst these patients are highly variable but when devices fail patients experience loss of voice and an increase risk of chest infection. Early failure of VP is a current clinical concern that is associated with regular hospital visits, reduced quality of life and associated medical cost. Poly-microbial biofilms comprised of both bacterial and fungal microorganisms readily colonize VPs and are linked to loss of device performance and its early failure in addition to providing a reservoir for potential infection. Our detailed analysis of poly-microbial biofilm composition on 159 early failing VPs from 48 total laryngectomy patients confirmed Candida albicans as the predominant fungal species and Staphylococcus aureus as the most common bacterial colonizer within our patient cohort. Using a combination of microbiological analysis, patient data and a high-throughput antifungal test assay mimicking in vivo conditions we established an evidence based precision antifungal treatment approach to VP management. Our approach has allowed us to implement a personalized VP management pathway, which increases device in situ lifespan by an average of 270%. Our study represents a significant step forward in both our understanding of the cause of VP failure and a new effective treatment pathway that offers tangible benefit to patients.

13.
Clin Cancer Res ; 25(21): 6406-6416, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31371342

RESUMEN

PURPOSE: OX40 agonist-based combinations are emerging as a novel avenue to improve the effectiveness of cancer immunotherapy. To better guide its clinical development, we characterized the role of the OX40 pathway in tumor-reactive immune cells. We also evaluated combining OX40 agonists with targeted therapy to combat resistance to cancer immunotherapy.Experimental Design: We utilized patient-derived tumor-infiltrating lymphocytes (TILs) and multiple preclinical models to determine the direct effect of anti-OX40 agonistic antibodies on tumor-reactive CD8+ T cells. We also evaluated the antitumor activity of an anti-OX40 antibody plus PI3Kß inhibition in a transgenic murine melanoma model (Braf mutant, PTEN null), which spontaneously develops immunotherapy-resistant melanomas. RESULTS: We observed elevated expression of OX40 in tumor-reactive CD8+ TILs upon encountering tumors; activation of OX40 signaling enhanced their cytotoxic function. OX40 agonist antibody improved the antitumor activity of CD8+ T cells and the generation of tumor-specific T-cell memory in vivo. Furthermore, combining anti-OX40 with GSK2636771, a PI3Kß-selective inhibitor, delayed tumor growth and extended the survival of mice with PTEN-null melanomas. This combination treatment did not increase the number of TILs, but it instead significantly enhanced proliferation of CD8+ TILs and elevated the serum levels of CCL4, CXCL10, and IFNγ, which are mainly produced by memory and/or effector T cells. CONCLUSIONS: These results highlight a critical role of OX40 activation in potentiating the effector function of tumor-reactive CD8+ T cells and suggest further evaluation of OX40 agonist-based combinations in patients with immune-resistant tumors.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Melanoma/tratamiento farmacológico , Fosfohidrolasa PTEN/genética , Receptores OX40/inmunología , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Inmunoterapia , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Ratones , Receptores OX40/antagonistas & inhibidores
14.
Nat Commun ; 9(1): 3919, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254248

RESUMEN

In addition to genomic mutations, RNA editing is another major mechanism creating sequence variations in proteins by introducing nucleotide changes in mRNA sequences. Deregulated RNA editing contributes to different types of human diseases, including cancers. Here we report that peptides generated as a consequence of RNA editing are indeed naturally presented by human leukocyte antigen (HLA) molecules. We provide evidence that effector CD8+ T cells specific for edited peptides derived from cyclin I are present in human tumours and attack tumour cells that are presenting these epitopes. We show that subpopulations of cancer patients have increased peptide levels and that levels of edited RNA correlate with peptide copy numbers. These findings demonstrate that RNA editing extends the classes of HLA presented self-antigens and that these antigens can be recognised by the immune system.


Asunto(s)
Antígenos de Neoplasias/inmunología , Epítopos/inmunología , Sistema Inmunológico/inmunología , Neoplasias/inmunología , Edición de ARN/inmunología , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Células Cultivadas , Ciclina I/genética , Ciclina I/inmunología , Ciclina I/metabolismo , Citotoxicidad Inmunológica/inmunología , Antígenos HLA/inmunología , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Péptidos/genética , Péptidos/inmunología , Péptidos/metabolismo , Proteogenómica/métodos
15.
Cell Metab ; 27(5): 977-987.e4, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29628419

RESUMEN

Adoptive T cell therapy (ACT) produces durable responses in some cancer patients; however, most tumors are refractory to ACT and the molecular mechanisms underlying resistance are unclear. Using two independent approaches, we identified tumor glycolysis as a pathway associated with immune resistance in melanoma. Glycolysis-related genes were upregulated in melanoma and lung cancer patient samples poorly infiltrated by T cells. Overexpression of glycolysis-related molecules impaired T cell killing of tumor cells, whereas inhibition of glycolysis enhanced T cell-mediated antitumor immunity in vitro and in vivo. Moreover, glycolysis-related gene expression was higher in melanoma tissues from ACT-refractory patients, and tumor cells derived from these patients exhibited higher glycolytic activity. We identified reduced levels of IRF1 and CXCL10 immunostimulatory molecules in highly glycolytic melanoma cells. Our findings demonstrate that tumor glycolysis is associated with the efficacy of ACT and identify the glycolysis pathway as a candidate target for combinatorial therapeutic intervention.


Asunto(s)
Glucólisis , Inmunoterapia Adoptiva , Neoplasias Pulmonares/terapia , Melanoma/terapia , Linfocitos T/trasplante , Animales , Línea Celular Tumoral , Quimiocina CXCL10/metabolismo , Femenino , Humanos , Factor 1 Regulador del Interferón/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Masculino , Melanoma/inmunología , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL
16.
Clin Cancer Res ; 24(14): 3366-3376, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29496759

RESUMEN

Purpose: Cancer immunotherapy has shown promising clinical outcomes in many patients. However, some patients still fail to respond, and new strategies are needed to overcome resistance. The purpose of this study was to identify novel genes and understand the mechanisms that confer resistance to cancer immunotherapy.Experimental Design: To identify genes mediating resistance to T-cell killing, we performed an open reading frame (ORF) screen of a kinome library to study whether overexpression of a gene in patient-derived melanoma cells could inhibit their susceptibility to killing by autologous tumor-infiltrating lymphocytes (TIL).Results: The RNA-binding protein MEX3B was identified as a top candidate that decreased the susceptibility of melanoma cells to killing by TILs. Further analyses of anti-PD-1-treated melanoma patient tumor samples suggested that higher MEX3B expression is associated with resistance to PD-1 blockade. In addition, significantly decreased levels of IFNγ were secreted from TILs incubated with MEX3B-overexpressing tumor cells. Interestingly, this phenotype was rescued upon overexpression of exogenous HLA-A2. Consistent with this, we observed decreased HLA-A expression in MEX3B-overexpressing tumor cells. Finally, luciferase reporter assays and RNA-binding protein immunoprecipitation assays suggest that this is due to MEX3B binding to the 3' untranslated region (UTR) of HLA-A to destabilize the mRNA.Conclusions: MEX3B mediates resistance to cancer immunotherapy by binding to the 3' UTR of HLA-A to destabilize the HLA-A mRNA and thus downregulate HLA-A expression on the surface of tumor cells, thereby making the tumor cells unable to be recognized and killed by T cells. Clin Cancer Res; 24(14); 3366-76. ©2018 AACRSee related commentary by Kalbasi and Ribas, p. 3239.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Antígenos HLA-A/genética , Proteínas de Unión al ARN/genética , Regiones no Traducidas 3' , Biomarcadores de Tumor , Línea Celular Tumoral , Citotoxicidad Inmunológica/genética , Genes Reporteros , Antígenos HLA-A/inmunología , Antígenos HLA-A/metabolismo , Humanos , Interferón gamma/biosíntesis , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Unión Proteica , Proteínas de Unión al ARN/metabolismo
17.
J Natl Cancer Inst ; 110(7): 777-786, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29267866

RESUMEN

Background: Immunotherapy has increasingly become a staple in cancer treatment. However, substantial limitations in the durability of response highlight the need for more rational therapeutic combinations. The aim of this study is to investigate how to make tumor cells more sensitive to T-cell-based cancer immunotherapy. Methods: Two pairs of melanoma patient-derived tumor cell lines and their autologous tumor-infiltrating lymphocytes were utilized in a high-throughput screen of 850 compounds to identify bioactive agents that could be used in combinatorial strategies to improve T-cell-mediated killing of tumor cells. RNAi, overexpression, and gene expression analyses were utilized to identify the mechanism underlying the effect of Topoisomerase I (Top1) inhibitors on T-cell-mediated killing. Using a syngeneic mouse model (n = 5 per group), the antitumor efficacy of the combination of a clinically relevant Top1 inhibitor, liposomal irinotecan (MM-398), with immune checkpoint inhibitors was also assessed. All statistical tests were two-sided. Results: We found that Top1 inhibitors increased the sensitivity of patient-derived melanoma cell lines (n = 7) to T-cell-mediated cytotoxicity (P < .001, Dunnett's test). This enhancement is mediated by TP53INP1, whose overexpression increased the susceptibility of melanoma cell lines to T-cell cytotoxicity (2549 cell line: P = .009, unpaired t test), whereas its knockdown impeded T-cell killing of Top1 inhibitor-treated melanoma cells (2549 cell line: P < .001, unpaired t test). In vivo, greater tumor control was achieved with MM-398 in combination with α-PD-L1 or α-PD1 (P < .001, Tukey's test). Prolonged survival was also observed in tumor-bearing mice treated with MM-398 in combination with α-PD-L1 (P = .002, log-rank test) or α-PD1 (P = .008, log-rank test). Conclusions: We demonstrated that Top1 inhibitors can improve the antitumor efficacy of cancer immunotherapy, thus providing the basis for developing novel strategies using Top1 inhibitors to augment the efficacy of immunotherapy.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Melanoma/terapia , Linfocitos T Citotóxicos/trasplante , Inhibidores de Topoisomerasa I/uso terapéutico , Animales , Línea Celular Tumoral , Quimioterapia Adyuvante , Terapia Combinada , Femenino , Humanos , Irinotecán/uso terapéutico , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/trasplante , Melanoma/inmunología , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos/inmunología , Topotecan/uso terapéutico , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nat Commun ; 8(1): 451, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28878208

RESUMEN

T-cell-based immunotherapies are promising treatments for cancer patients. Although durable responses can be achieved in some patients, many patients fail to respond to these therapies, underscoring the need for improvement with combination therapies. From a screen of 850 bioactive compounds, we identify HSP90 inhibitors as candidates for combination with immunotherapy. We show that inhibition of HSP90 with ganetespib enhances T-cell-mediated killing of patient-derived human melanoma cells by their autologous T cells in vitro and potentiates responses to anti-CTLA4 and anti-PD1 therapy in vivo. Mechanistic studies reveal that HSP90 inhibition results in upregulation of interferon response genes, which are essential for the enhanced killing of ganetespib treated melanoma cells by T cells. Taken together, these findings provide evidence that HSP90 inhibition can potentiate T-cell-mediated anti-tumor immune responses, and rationale to explore the combination of immunotherapy and HSP90 inhibitors.Many patients fail to respond to T cell based immunotherapies. Here, the authors, through a high-throughput screening, identify HSP90 inhibitors as a class of preferred drugs for treatment combination with immunotherapy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Ipilimumab/farmacología , Melanoma/terapia , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Inmunoterapia , Interferones/farmacología , Estimación de Kaplan-Meier , Melanoma/genética , Melanoma/metabolismo , Ratones Endogámicos C57BL , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Regulación hacia Arriba
19.
Cancer Discov ; 6(2): 202-16, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26645196

RESUMEN

UNLABELLED: T cell-mediated immunotherapies are promising cancer treatments. However, most patients still fail to respond to these therapies. The molecular determinants of immune resistance are poorly understood. We show that loss of PTEN in tumor cells in preclinical models of melanoma inhibits T cell-mediated tumor killing and decreases T-cell trafficking into tumors. In patients, PTEN loss correlates with decreased T-cell infiltration at tumor sites, reduced likelihood of successful T-cell expansion from resected tumors, and inferior outcomes with PD-1 inhibitor therapy. PTEN loss in tumor cells increased the expression of immunosuppressive cytokines, resulting in decreased T-cell infiltration in tumors, and inhibited autophagy, which decreased T cell-mediated cell death. Treatment with a selective PI3Kß inhibitor improved the efficacy of both anti-PD-1 and anti-CTLA-4 antibodies in murine models. Together, these findings demonstrate that PTEN loss promotes immune resistance and support the rationale to explore combinations of immunotherapies and PI3K-AKT pathway inhibitors. SIGNIFICANCE: This study adds to the growing evidence that oncogenic pathways in tumors can promote resistance to the antitumor immune response. As PTEN loss and PI3K-AKT pathway activation occur in multiple tumor types, the results support the rationale to further evaluate combinatorial strategies targeting the PI3K-AKT pathway to increase the efficacy of immunotherapy.


Asunto(s)
Anticuerpos/administración & dosificación , Melanoma/tratamiento farmacológico , Melanoma/genética , Fosfohidrolasa PTEN/deficiencia , Linfocitos T/inmunología , Aminopiridinas/administración & dosificación , Aminopiridinas/uso terapéutico , Animales , Anticuerpos/uso terapéutico , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Inmunoterapia/métodos , Melanoma/inmunología , Ratones , Morfolinas/administración & dosificación , Morfolinas/uso terapéutico , Receptor de Muerte Celular Programada 1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...