Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Cancer Immunol Res ; 11(10): 1314-1331, 2023 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-37540803

RESUMEN

Infiltration of tumor by T cells is a prerequisite for successful immunotherapy of solid tumors. In this study, we investigate the influence of tumor-targeted radiation on chimeric antigen receptor (CAR) T-cell therapy tumor infiltration, accumulation, and efficacy in clinically relevant models of pleural mesothelioma and non-small cell lung cancers. We use a nonablative dose of tumor-targeted radiation prior to systemic administration of mesothelin-targeted CAR T cells to assess infiltration, proliferation, antitumor efficacy, and functional persistence of CAR T cells at primary and distant sites of tumor. A tumor-targeted, nonablative dose of radiation promotes early and high infiltration, proliferation, and functional persistence of CAR T cells. Tumor-targeted radiation promotes tumor-chemokine expression and chemokine-receptor expression in infiltrating T cells and results in a subpopulation of higher-intensity CAR-expressing T cells with high coexpression of chemokine receptors that further infiltrate distant sites of disease, enhancing CAR T-cell antitumor efficacy. Enhanced CAR T-cell efficacy is evident in models of both high-mesothelin-expressing mesothelioma and mixed-mesothelin-expressing lung cancer-two thoracic cancers for which radiotherapy is part of the standard of care. Our results strongly suggest that the use of tumor-targeted radiation prior to systemic administration of CAR T cells may substantially improve CAR T-cell therapy efficacy for solid tumors. Building on our observations, we describe a translational strategy of "sandwich" cell therapy for solid tumors that combines sequential metastatic site-targeted radiation and CAR T cells-a regional solution to overcome barriers to systemic delivery of CAR T cells.


Asunto(s)
Mesotelioma Maligno , Mesotelioma , Humanos , Mesotelina , Inmunoterapia Adoptiva/métodos , Proteínas Ligadas a GPI , Receptores de Antígenos de Linfocitos T , Mesotelioma/radioterapia , Mesotelioma Maligno/tratamiento farmacológico , Receptores de Quimiocina , Quimiocinas , Línea Celular Tumoral
3.
Nat Cancer ; 4(7): 1001-1015, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37336986

RESUMEN

The limited efficacy of chimeric antigen receptor (CAR) T cell therapy for solid tumors necessitates engineering strategies that promote functional persistence in an immunosuppressive environment. Herein, we use c-Kit signaling, a physiological pathway associated with stemness in hematopoietic progenitor cells (T cells lose expression of c-Kit during differentiation). CAR T cells with intracellular expression, but no cell-surface receptor expression, of the c-Kit D816V mutation (KITv) have upregulated STAT phosphorylation, antigen activation-dependent proliferation and CD28- and interleukin-2-independent and interferon-γ-mediated co-stimulation, augmenting the cytotoxicity of first-generation CAR T cells. This translates to enhanced survival, including in transforming growth factor-ß-rich and low-antigen-expressing solid tumor models. KITv CAR T cells have equivalent or better in vivo efficacy than second-generation CAR T cells and are susceptible to tyrosine kinase inhibitors (safety switch). When combined with CD28 co-stimulation, KITv co-stimulation functions as a third signal, enhancing efficacy and providing a potent approach to treat solid tumors.


Asunto(s)
Interleucina-2 , Proteínas Proto-Oncogénicas c-kit , Linfocitos T , Antígenos CD28/genética , Antígenos CD28/metabolismo , Línea Celular Tumoral , Inmunoterapia Adoptiva , Interleucina-2/farmacología , Interleucina-2/metabolismo , Proteínas Tirosina Quinasas Receptoras , Proteínas Proto-Oncogénicas c-kit/metabolismo
4.
Expert Opin Investig Drugs ; 31(11): 1187-1202, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36448335

RESUMEN

INTRODUCTION: In solid tumor immunotherapy, less than 20% of patients respond to anti-programmed cell death 1 (PD-1)/programmed cell death 1 ligand 1 (PD-L1) agents. The role of transforming growth factor ß (TGFß) in diverse immunity is well-established; however, systemic blockade of TGFß is associated with toxicity. Accumulating evidence suggests the role of crosstalk between TGFß and PD-1/PD-L1 pathways. AREAS COVERED: We focus on TGFß and PD-1/PD-L1 signaling pathway crosstalk and the determinant role of TGFß in the resistance of immune checkpoint blockade. We provide the rationale for combination anti-TGFß and anti-PD-1/PD-L1 therapies for solid tumors and discuss the current status of dual blockade therapy in preclinical and clinical studies. EXPERT OPINION: The heterogeneity of tumor microenvironment across solid tumors complicates patient selection, treatment regimens, and response and toxicity assessment for investigation of dual blockade agents. However, clinical knowledge from single-agent studies provides infrastructure to translate dual blockade therapies. Dual TGFß and PD-1/PD-L1 blockade results in enhanced T-cell infiltration into tumors, a primary requisite for successful immunotherapy. A bifunctional fusion protein specifically targets TGFß in the tumor microenvironment, avoiding systemic toxicity, and prevents interaction of PD-1+ cytotoxic cells with PD-L1+ tumor cells.


Asunto(s)
Neoplasias , Factor de Crecimiento Transformador beta , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Antígeno B7-H1 , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Transducción de Señal , Microambiente Tumoral
5.
Cancer Cell ; 40(6): 569-574, 2022 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-35487216

RESUMEN

The success of chimeric antigen receptor (CAR) T cell therapy in solid tumors, unlike in hematologic malignancies, is limited by inadequate tumor infiltration and T cell dysfunction and exhaustion. Regional delivery of CAR T cells in patients with solid tumors is safe and feasible; promotes infiltration, proliferation, and trafficking; and ignites functionally persisting systemic immunity.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Antígenos de Neoplasias , Humanos , Inmunoterapia Adoptiva , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T , Microambiente Tumoral
6.
Viruses ; 13(8)2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34452350

RESUMEN

Persistent infections with some types of human papillomavirus (HPV) constitute the major etiological factor for cervical cancer development. Nanog, a stem cell transcription factor has been shown to increase during cancer progression. We wanted to determine whether Nanog could modulate transcription of E6 and E7 oncogenes. We used luciferase reporters under the regulation of the long control region (LCR) of HPV types 16 and 18 (HPV16/18) and performed RT-qPCR. We found that Nanog increases activity of both viral regulatory regions and elevates endogenous E6/E7 mRNA levels in cervical cancer-derived cells. We demonstrated by in vitro mutagenesis that changes at Nanog-binding sites found in the HPV18 LCR significantly inhibit transcriptional activation. Chromatin immunoprecipitation (ChIP) assays showed that Nanog binds in vivo to the HPV18 LCR, and its overexpression increases its binding as well as that of c-Jun. Surprisingly, we observed that mutation of AP1-binding sites also affect Nanog's ability to activate transcription, suggesting cooperation between the two factors. We searched for putative Nanog-binding sites in the LCR of several HPVs and surprisingly found them only in those types associated with cancer development. Our study shows, for the first time, a role for Nanog in the regulation of E6/E7 transcription of HPV16/18.


Asunto(s)
Proteínas de Unión al ADN/genética , Papillomavirus Humano 16/genética , Papillomavirus Humano 18/genética , Proteína Homeótica Nanog/metabolismo , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus/genética , Infecciones por Papillomavirus/metabolismo , Proteínas Represoras/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 18/metabolismo , Humanos , Proteína Homeótica Nanog/genética , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/virología , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/virología
7.
Viral Immunol ; 32(6): 269-275, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31199716

RESUMEN

CD8+ T cell immune response plays a critical role in the clearance of human papillomavirus (HPV)-infected cells. During the natural history of HPV infection, the E1 protein, an early-expressed helicase highly conserved among papillomaviruses, is involved in the replication of HPV genomes. We have previously shown, in a murine model, that immunization with HPV18 E1 protein combined with α-galactosylceramide elicits a specific CD8+ T cell response. We further proved those findings by analyzing whether CD8+ T cells from mice immunized with α-galactosylceramide plus HPV18 E1 protein could have a cytotoxic effect on cells expressing the carboxyl-terminal domain from the E1 proteins of other HPV types. Interestingly, CD8+ T cells raised against HPV18 E1 antigen presented cross-reactivity against the E1 protein from HPV53, 33, 16, and 31. Poor cross-reactivity was observed for HPV11, and none for HPV6. This outcome may be relevant for the design of broad-spectrum immune-protective agents against HPV infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Reacciones Cruzadas , Galactosilceramidas/inmunología , Proteínas Oncogénicas Virales/inmunología , Animales , Pruebas Inmunológicas de Citotoxicidad , Femenino , Galactosilceramidas/administración & dosificación , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunización , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/administración & dosificación , Papillomaviridae/clasificación , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/prevención & control , Bazo/citología , Bazo/inmunología
8.
Vaccine ; 37(9): 1219-1228, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30704821

RESUMEN

CD8+ T cell-mediated immune response plays a major role in the clearance of virus-infected cells, including human papillomavirus (HPV). The effective treatment of women with normal cytology but persistent high risk-HPV infection or with low-grade intraepithelial lesions could take advantage of novel strategies based on vaccination with viral immunological targets with a wide spectrum of cross-protection. The helicase E1, expressed early during viral replication in HPV infection, is among the most conserved papillomavirus proteins, which makes it a good vaccine candidate. In the present study, we examined E1-specific CD8+ T cell and NK immune responses in a mouse model with α-galactosylceramide (α-GalCer) as an adjuvant. We found that mice immunized with E1 combined with α-GalCer elicited an E1-specific CD8+ T and NK cell cytotoxic responses, which correlated with growth inhibition of grafted melanoma B16-F0 cells expressing E1, both in prophylactic and therapeutic protocols.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Citotoxicidad Inmunológica , Galactosilceramidas/administración & dosificación , Proteínas Oncogénicas Virales/inmunología , Linfocitos T Citotóxicos/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Femenino , Galactosilceramidas/inmunología , Papillomavirus Humano 18 , Humanos , Células Asesinas Naturales/inmunología , Melanoma Experimental/prevención & control , Melanoma Experimental/terapia , Melanoma Experimental/virología , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/administración & dosificación , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/terapia , Trasplantes , Células Tumorales Cultivadas/inmunología , Vacunación
9.
Cancers (Basel) ; 10(11)2018 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-30373251

RESUMEN

Oncolytic virotherapy has been investigated for several decades and is emerging as a plausible biological therapy with several ongoing clinical trials and two viruses are now approved for cancer treatment in humans. The direct cytotoxicity and immune-stimulatory effects make oncolytic viruses an interesting strategy for cancer treatment. In this review, we summarize the results of in vitro and in vivo published studies of oncolytic viruses in different phases of evaluation in dogs, using PubMed and Google scholar as search platforms, without time restrictions (to date). Natural and genetically modified oncolytic viruses were evaluated with some encouraging results. The most studied viruses to date are the reovirus, myxoma virus, and vaccinia, tested mostly in solid tumors such as osteosarcomas, mammary gland tumors, soft tissue sarcomas, and mastocytomas. Although the results are promising, there are issues that need addressing such as ensuring tumor specificity, developing optimal dosing, circumventing preexisting antibodies from previous exposure or the development of antibodies during treatment, and assuring a reasonable safety profile, all of which are required in order to make this approach a successful therapy in dogs.

10.
Viruses ; 9(7)2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28678184

RESUMEN

Persistent infections with high-risk human papillomavirus (HPV) constitute the main risk factor for cervical cancer development. HPV16 is the most frequent type associated to squamous cell carcinomas (SCC), followed by HPV18. The long control region (LCR) in the HPV genome contains the replication origin and sequences recognized by cellular transcription factors (TFs) controlling viral transcription. Altered expression of E6 and E7 viral oncogenes, modulated by the LCR, causes modifications in cellular pathways such as proliferation, leading to malignant transformation. The aim of this study was to identify specific TFs that could contribute to the modulation of high-risk HPV transcriptional activity, related to the cellular histological origin. We identified sex determining region Y (SRY)-box 2 (SOX2) response elements present in HPV16-LCR. SOX2 binding to the LCR was demonstrated by in vivo and in vitro assays. The overexpression of this TF repressed HPV16-LCR transcriptional activity, as shown through reporter plasmid assays and by the down-regulation of endogenous HPV oncogenes. Site-directed mutagenesis revealed that three putative SOX2 binding sites are involved in the repression of the LCR activity. We propose that SOX2 acts as a transcriptional repressor of HPV16-LCR, decreasing the expression of E6 and E7 oncogenes in a SCC context.


Asunto(s)
Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Papillomavirus Humano 16/fisiología , Proteínas Oncogénicas Virales/biosíntesis , Factores de Transcripción SOXB1/metabolismo , Transcripción Genética , Sitios de Unión , Línea Celular , Análisis Mutacional de ADN , Papillomavirus Humano 16/genética , Humanos , Mutagénesis Sitio-Dirigida , Unión Proteica
11.
BMC Cancer ; 13: 445, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24083491

RESUMEN

BACKGROUND: Metaplastic carcinoma, an uncommon subtype of breast cancer, is part of the spectrum of basal-like, triple receptor-negative breast carcinomas. The present study examined 20 surgical specimens of metaplastic breast carcinomas, for the presence of high-risk Human papillomavirus (HPV), which is suspected to be a potential carcinogenic agent for breast carcinoma. METHODS: Mastectomy specimens from patients harboring metaplastic breast carcinoma, as defined by the World Health Organization (WHO), and who attended the Instituto Nacional de Cancerologia in Mexico City, were retrieved from the files of the Department of Pathology accumulated during a 16-year period (1995-2008). Demographic and clinical information was obtained from patients' medical records. DNA was extracted from formalin-fixed, paraffin-embedded tumors and HPV type-specific amplification was performed by means of Polymerase chain reaction (PCR). Quantitative Real-time (RT) PCR was conducted in HPV positive cases. Statistically, the association of continuous or categorical variables with HPV status was tested by the Student t, the Chi square, or Fisher's exact tests, as appropriate. RESULTS: High-risk HPV DNA was detected in eight (40%) of 20 metaplastic breast carcinomas: seven (87.5%) HPV-16 and one (12.5%) HPV-18. Mean age of patients with HPV-positive cases was 49 years (range 24-72 years), the same as for HPV-negative cases (range, 30-73 years). There were not striking differences between HPV + and HPV- metaplastic carcinomas regarding clinical findings. Nearly all cases were negative for estrogen, progesterone and Human epidermal growth factor receptor 2 (HER2), but positive for Epidermal growth factor receptor (EGFR). CONCLUSIONS: High-risk HPV has been strongly associated with conventional breast carcinomas, although the subtle mechanism of neoplastic transformation is poorly understood. In Mexican patients, the prevalence of HPV infection among metaplastic breast carcinomas is higher than in non-metaplastic ones, as so the HPV viral loads; notwithstanding, HPV viral loads show wide variation and remain even lower than cervical and other non-cervical carcinomas, making it difficult to assume that HPV could play a key role in breast carcinogenesis. Further studies are warranted to elucidate the meaning of the presence of high-risk HPVDNA in breast carcinomas.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias de la Mama/virología , ADN Viral , Papillomaviridae/genética , Infecciones por Papillomavirus/virología , Adulto , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/epidemiología , Femenino , Dosificación de Gen , Papillomavirus Humano 16/genética , Papillomavirus Humano 18/genética , Humanos , México , Persona de Mediana Edad , Clasificación del Tumor , Metástasis de la Neoplasia , Estadificación de Neoplasias , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/epidemiología , Receptores de Estrógenos/metabolismo , Recurrencia , Carga Tumoral
12.
Viruses ; 5(11): 2624-42, 2013 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-24169630

RESUMEN

During the early stages of human papillomavirus (HPV) infections, the innate immune system creates a pro-inflammatory microenvironment by recruiting innate immune cells to eliminate the infected cells, initiating an effective acquired immune response. However, HPV exhibits a wide range of strategies for evading immune-surveillance, generating an anti-inflammatory microenvironment. The administration of new adjuvants, such as TLR (Toll-like receptors) agonists and alpha-galactosylceramide, has been demonstrated to reverse the anti-inflammatory microenvironment by down-regulating a number of adhesion molecules and chemo-attractants and activating keratinocytes, dendritic (DC), Langerhans (LC), natural killer (NK) or natural killer T (NKT) cells; thus, promoting a strong specific cytotoxic T cell response. Therefore, these adjuvants show promise for the treatment of HPV generated lesions and may be useful to elucidate the unknown roles of immune cells in the natural history of HPV infection. This review focuses on HPV immune evasion mechanisms and on the proposed response of the innate immune system, suggesting a role for the surrounding pro-inflammatory microenvironment and the NK and NKT cells in the clearance of HPV infections.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Inmunidad Innata , Papillomaviridae/inmunología , Infecciones por Papillomavirus/tratamiento farmacológico , Infecciones por Papillomavirus/inmunología , Animales , Humanos , Evasión Inmune , Inmunidad Innata/efectos de los fármacos , Papillomaviridae/genética , Papillomaviridae/fisiología , Infecciones por Papillomavirus/virología
13.
J Gen Virol ; 94(Pt 2): 393-402, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23100366

RESUMEN

A persistent infection with high-risk human papillomavirus (HPV) constitutes the main aetiological factor for cervical cancer development. HPV16 and 18 are the most prevalent types found in cervical cancer worldwide. It has been proposed that HPV intratype variations may result in differences in biological behaviour. Three different HPV18 variants belonging to the Asian Amerindian (AsAi), European (E) and African (Af) branches have been associated with specific histological types of cervical cancer with different relative prognoses, suggesting that HPV18 genomic variations might participate in disease evolution. The E1 viral protein plays a critical role in controlling viral replication and load, requiring interaction with the E2 protein to bind to the long control region (LCR). In this work, we analysed if intratype variations in the LCR and E1 and E2 genes of HPV18 impact ori replication. While the changes found in E2 genes of the tested variants were irrelevant in replication, we found that variations in E1 and LCR in fact affect ori function. It was demonstrated that nucleotide differences in the LCR variants impact ori function. Nevertheless, HPV18 E1 Af gene was mainly involved in the highest ori replication, compared with the E and AsAi E1 variants. Immunofluorescence analysis showed increased levels of Af E1 in the nucleus, correlating with the enhanced ori function. Site-directed mutagenesis revealed that at least two positions in the N-terminal domain of E1 could impact its nuclear accumulation.


Asunto(s)
Papillomavirus Humano 18/genética , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Regiones Promotoras Genéticas , Replicación Viral , Núcleo Celular/metabolismo , Núcleo Celular/virología , Papillomavirus Humano 18/fisiología , Humanos
14.
Rev Invest Clin ; 62(1): 81-90, 2010.
Artículo en Español | MEDLINE | ID: mdl-20415062

RESUMEN

Small RNAs belong to a newly discovered strain of molecules. These molecules are composed of double strand RNA comprised by just about 19-31 nucleotides. They have two main characteristics that make them unique. Firstly, they are noncoding for proteins and second they interfere post-transcriptional with mRNA. This interfering action is the distinguishing hallmark, therefore known as interfering RNA or RNAi. There are three main subclasses of which micro-RNA and siRNA are the most widely studied. Interference RNAs participate in a myriad of cellular functions mainly through modulation of genetic expression. Due to these capabilities it has been used as therapeutic weapon in a number of diseases including cancer. It is known that both miRNA and siRNA participate in carcinogenesis, either inhibiting suppressor genes, or stimulating oncogenes. It has been demonstrated that manipulating small interfering RNAs in cell lines and animal models, the malignant and metastatic phenotype can be reversed. Up to now a few clinical trials using RNAi as a therapeutic agent have demonstrated some success and feasibility. It is forseeable that in the near future cancer treatment with small RNAs will be widely applicable, once the many constrains for its systemic application are surpassed.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Terapia Genética/métodos , MicroARNs/genética , Neoplasias/genética , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/patología , Transformación Celular Neoplásica/genética , Ensayos Clínicos como Asunto , Sistemas de Liberación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Estudios de Factibilidad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor , Vectores Genéticos/uso terapéutico , Humanos , Masculino , MicroARNs/biosíntesis , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Neoplasias/virología , Oncogenes , ARN Mensajero/antagonistas & inhibidores , ARN Neoplásico/antagonistas & inhibidores , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , ARN Interferente Pequeño/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...