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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 22(8): 1042-1051, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34267375

RESUMEN

Pathogens and vaccines that produce persisting antigens can generate expanded pools of effector memory CD8+ T cells, described as memory inflation. While properties of inflating memory CD8+ T cells have been characterized, the specific cell types and tissue factors responsible for their maintenance remain elusive. Here, we show that clinically applied adenovirus vectors preferentially target fibroblastic stromal cells in cultured human tissues. Moreover, we used cell-type-specific antigen targeting to define critical cells and molecules that sustain long-term antigen presentation and T cell activity after adenovirus vector immunization in mice. While antigen targeting to myeloid cells was insufficient to activate antigen-specific CD8+ T cells, genetic activation of antigen expression in Ccl19-cre-expressing fibroblastic stromal cells induced inflating CD8+ T cells. Local ablation of vector-targeted cells revealed that lung fibroblasts support the protective function and metabolic fitness of inflating memory CD8+ T cells in an interleukin (IL)-33-dependent manner. Collectively, these data define a critical fibroblastic niche that underpins robust protective immunity operating in a clinically important vaccine platform.


Asunto(s)
Adenoviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Interleucina-33/inmunología , Activación de Linfocitos/inmunología , Células del Estroma/inmunología , Adenoviridae/genética , Animales , Línea Celular Tumoral , Quimiocina CCL19/metabolismo , Quimera/genética , Epítopos de Linfocito T/inmunología , Fibroblastos/citología , Fibroblastos/metabolismo , Vectores Genéticos/inmunología , Humanos , Pulmón/citología , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vacunación
2.
Mol Ther ; 32(2): 426-439, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38058126

RESUMEN

Harnessing the immune system to eradicate tumors requires identification and targeting of tumor antigens, including tumor-specific neoantigens and tumor-associated self-antigens. Tumor-associated antigens are subject to existing immune tolerance, which must be overcome by immunotherapies. Despite many novel immunotherapies reaching clinical trials, inducing self-antigen-specific immune responses remains challenging. Here, we systematically investigate viral-vector-based cancer vaccines encoding a tumor-associated self-antigen (TRP2) for the treatment of established melanomas in preclinical mouse models, alone or in combination with adoptive T cell therapy. We reveal that, unlike foreign antigens, tumor-associated antigens require replication of lymphocytic choriomeningitis virus (LCMV)-based vectors to break tolerance and induce effective antigen-specific CD8+ T cell responses. Immunization with a replicating LCMV vector leads to complete tumor rejection when combined with adoptive TRP2-specific T cell transfer. Importantly, immunization with replicating vectors leads to extended antigen persistence in secondary lymphoid organs, resulting in efficient T cell priming, which renders previously "cold" tumors open to immune infiltration and reprograms the tumor microenvironment to "hot." Our findings have important implications for the design of next-generation immunotherapies targeting solid cancers utilizing viral vectors and adoptive cell transfer.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Ratones , Animales , Virus de la Coriomeningitis Linfocítica/genética , Linfocitos T CD8-positivos , Neoplasias/tratamiento farmacológico , Antígenos de Neoplasias/genética , Autoantígenos , Microambiente Tumoral
3.
Cancer Immunol Immunother ; 70(2): 563-568, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32804246

RESUMEN

Immune checkpoint inhibitors (ICIs) are emerging as the new standard of care for treating various metastatic cancers. It is known that effective anti-tumor immune responses are associated with a stronger presence of tumor-infiltrating lymphocytes (TILs) in solid tumor tissue. Cancer patients with relapsing-remitting multiple sclerosis (RRMS) are often under continuous treatment with fingolimod, an immune-modulating drug that inhibits lymphocyte egress from secondary lymphatic organs. Little is known about the effect of fingolimod on ICI cancer therapy, as fingolimod may limit the number of TILs. Here we present three patients with RRMS, who developed various cancers during fingolimod treatment. Histology of all tumors consistently showed low numbers of TILs. A second biopsy taken from one of the tumors, a melanoma, revealed a significant increase of TILs after stopping fingolimod and starting pembrolizumab, indicating a surge in the number and re-invigoration of T cells. Our study suggests that fingolimod limits the number of TILs in solid tumors and may, thus, inhibit anti-cancer immune responses.


Asunto(s)
Clorhidrato de Fingolimod/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunosupresores/uso terapéutico , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos/inmunología , Neoplasias/tratamiento farmacológico , Femenino , Clorhidrato de Fingolimod/farmacología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunosupresores/farmacología , Persona de Mediana Edad , Neoplasias/inmunología
4.
J Am Acad Dermatol ; 82(4): 854-861, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31449902

RESUMEN

BACKGROUND: Anti-programmed cell death protein 1 (PD1)/programmed death-ligand 1(PD-L1) therapy frequently entails immune-related adverse events (irAEs), and biomarkers to predict irAEs are lacking. Although checkpoint inhibitors have been found to reinvigorate T cells, the relevance of autoantibodies remains elusive. OBJECTIVE: Our aim was to explore whether IgG autoantibodies directed against coexpressed antigens by tumor tissue and healthy skin correlate with skin irAEs and therapy outcome. METHODS: We measured skin-specific IgG via enzyme-linked immunosorbent assay in patients with non-small cell lung cancer (NSCLC) who received anti-PD1/PD-L1 treatment between July 2015 and September 2017 at the Kantonsspital St. Gallen. Sera were sampled at baseline and during therapy after 8 weeks. RESULTS: Analysis of publicly available tumor expression data revealed that NSCLC and skin coexpress BP180, BP230, and type VII collagen. A skin irAE developed in 16 of 40 recruited patients (40%). Only elevated anti-BP180 IgG at baseline significantly correlated with the development of skin irAEs (P = .04), therapy response (P = .01), and overall survival (P = .04). LIMITATIONS: The patients were recruited in a single tertiary care center. CONCLUSIONS: Our data suggest that the level of anti-BP180 IgG of NSCLC patients at baseline is associated with better therapy response and overall survival and with a higher probability to develop skin irAEs during anti-PD1/PD-L1 treatment.


Asunto(s)
Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/uso terapéutico , Autoantígenos/inmunología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Erupciones por Medicamentos/etiología , Inmunoglobulina G/sangre , Neoplasias Pulmonares/tratamiento farmacológico , Colágenos no Fibrilares/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Anciano , Biomarcadores/sangre , Exantema/inducido químicamente , Femenino , Humanos , Masculino , Estudios Prospectivos , Prurito/inducido químicamente , Análisis de Supervivencia , Colágeno Tipo XVII
6.
Nat Commun ; 12(1): 4734, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34354077

RESUMEN

The tumor microenvironment (TME) is a complex amalgam of tumor cells, immune cells, endothelial cells and fibroblastic stromal cells (FSC). Cancer-associated fibroblasts are generally seen as tumor-promoting entity. However, it is conceivable that particular FSC populations within the TME contribute to immune-mediated tumor control. Here, we show that intratumoral treatment of mice with a recombinant lymphocytic choriomeningitis virus-based vaccine vector expressing a melanocyte differentiation antigen resulted in T cell-dependent long-term control of melanomas. Using single-cell RNA-seq analysis, we demonstrate that viral vector-mediated transduction reprogrammed and activated a Cxcl13-expressing FSC subset that show a pronounced immunostimulatory signature and increased expression of the inflammatory cytokine IL-33. Ablation of Il33 gene expression in Cxcl13-Cre-positive FSCs reduces the functionality of intratumoral T cells and unleashes tumor growth. Thus, reprogramming of FSCs by a self-antigen-expressing viral vector in the TME is critical for curative melanoma treatment by locally sustaining the activity of tumor-specific T cells.


Asunto(s)
Melanoma Experimental/terapia , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Fibroblastos Asociados al Cáncer/inmunología , Fibroblastos Asociados al Cáncer/patología , Técnicas de Reprogramación Celular/métodos , Quimiocina CXCL13/genética , Quimiocina CXCL13/inmunología , Femenino , Vectores Genéticos , Interleucina-33/deficiencia , Interleucina-33/genética , Interleucina-33/inmunología , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/inmunología , Virus de la Coriomeningitis Linfocítica/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células del Estroma/inmunología , Células del Estroma/patología , Linfocitos T/inmunología , Linfocitos T/patología , Microambiente Tumoral/inmunología
7.
Mol Ther Oncolytics ; 19: 179-187, 2020 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-33209978

RESUMEN

Cancer vaccination aims at inducing an adaptive immune response against tumor-derived antigens. In this study, we utilize recombinant human adenovirus serotype 5 (rAd5) and recombinant lymphocytic choriomeningitis virus (rLCMV)-based vectors expressing the melanocyte differentiation antigen gp100. In contrast to single or homologous vaccination, a heterologous prime boost vaccination starting with a rAd5-gp100 prime immunization followed by a rLCMV-gp100 boost injection induces a high magnitude of polyfunctional gp100-specific CD8+ T cells. Our data indicate that an optimal T cell induction is dependent on the order and interval of the vaccinations. A prophylactic prime boost vaccination with rAd5- and rLCMV-gp100 protects mice from a B16.F10 melanoma challenge. In the therapeutic setting, combination of the vaccination with low-dose cyclophosphamide showed a synergistic effect and significantly delayed tumor growth. Our findings suggest that heterologous viral vector prime boost immunizations can mediate tumor control in a mouse melanoma model.

8.
Sci Immunol ; 5(51)2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917792

RESUMEN

Fibroblastic reticular cells (FRCs) are stromal cells that actively promote the induction of immune responses by coordinating the interaction of innate and adaptive immune cells. However, whether and to which extent immune cell activation is determined by lymph node FRC reprogramming during acute viral infection has remained unexplored. Here, we genetically ablated expression of the type I interferon-α receptor (Ifnar) in Ccl19-Cre+ cells and found that sensing of type I interferon imprints an antiviral state in FRCs and thereby preserves myeloid cell composition in lymph nodes of naive mice. During localized lymphocytic choriomeningitis virus infection, IFNAR signaling precipitated profound phenotypic adaptation of all FRC subsets enhancing antigen presentation, chemokine-driven immune cell recruitment, and immune regulation. The IFNAR-dependent shift of all FRC subsets toward an immunostimulatory state reduced exhaustive CD8+ T cell activation. In sum, these results unveil intricate circuits underlying type I IFN sensing in lymph node FRCs that enable protective antiviral immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Fibroblastos/inmunología , Interferón Tipo I/inmunología , Coriomeningitis Linfocítica/inmunología , Células del Estroma/inmunología , Animales , Línea Celular , Interferón gamma/inmunología , Virus de la Coriomeningitis Linfocítica , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal , Factor de Necrosis Tumoral alfa/inmunología
9.
Clin Epigenetics ; 12(1): 94, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32586358

RESUMEN

BACKGROUND: PD-L1 (programmed cell death 1 ligand 1) expression in melanoma has been associated with a better response to anti-PD-1 (programmed cell death 1) therapy. However, patients with PD-L1-negative melanomas can respond to anti-PD-1 blockade, suggesting that the other PD-1 ligand, PD-L2 (programmed cell death 1 ligand 2), might also be relevant for efficacy of PD-1 inhibition. We investigated PD-L2 expression and methylation as a prognostic and predictive biomarker in melanoma. METHODS: DNA methylation at five CpG loci and gene expression of PD-L2 were evaluated with regard to survival in 470 melanomas from The Cancer Genome Atlas. PD-L2 promoter methylation in correlation with PD-L2 mRNA and protein expression was analyzed in human melanoma cell lines. Prognostic and predictive value of PD-L2 methylation was validated using quantitative methylation-specific PCR in a multicenter cohort of 129 melanoma patients receiving anti-PD-1 therapy. mRNA sequencing data of 121 melanoma patients receiving anti-PD-1 therapy provided by Liu et al. were analyzed for PD-L2 mRNA expression. RESULTS: We found significant correlations between PD-L2 methylation and mRNA expression levels in melanoma tissues and cell lines. Interferon-γ inducible PD-L2 protein expression correlated with PD-L2 promoter methylation in melanoma cells. PD-L2 DNA promoter hypomethylation and high mRNA expression were found to be strong predictors of prolonged overall survival. In pre-treatment melanoma samples from patients receiving anti-PD-1 therapy, low PD-L2 DNA methylation and high PD-L2 mRNA expression predicted longer progression-free survival. CONCLUSION: PD-L2 expression seems to be regulated via DNA promoter methylation. PD-L2 DNA methylation and mRNA expression may predict progression-free survival in melanoma patients receiving anti-PD-1 immunotherapy. Assessment of PD-L2 should be included in further clinical trials with anti-PD-1 antibodies.


Asunto(s)
Metilación de ADN/genética , Melanoma/genética , Proteína 2 Ligando de Muerte Celular Programada 1/genética , ARN Mensajero/genética , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular/metabolismo , Línea Celular/patología , Estudios de Cohortes , Islas de CpG/genética , Femenino , Expresión Génica/genética , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/métodos , Masculino , Melanoma/diagnóstico , Melanoma/tratamiento farmacológico , Melanoma/mortalidad , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Supervivencia sin Progresión , Neoplasias Cutáneas/patología
10.
EBioMedicine ; 59: 102962, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32861198

RESUMEN

BACKGROUND: The co-receptor lymphocyte-activation gene-3 (LAG3, LAG-3, CD223) is a potential target for immune checkpoint inhibition immunotherapies. However, little is known about the biological and clinical significance of LAG3 DNA methylation in melanoma and its microenvironment. METHODS: We evaluated LAG3 promoter and gene body methylation in a cohort of N = 470 melanoma patients obtained from The Cancer Genome Atlas (TCGA cohort), an independent cohort of N = 120 patients from the University Hospital Bonn, and in subsets of peripheral blood leukocytes, melanocytes, and melanoma cell lines. We validated the association of LAG3 methylation with mRNA expression in vitro in the melanoma cell line A375 treated with the hypomethylating agent 5-azacytidine and stimulated with interferon-γ. Finally, we investigated correlations between LAG3 methylation and progression-free survival in patients treated with immune checkpoint blockade (ICB cohort, N = 118). FINDINGS: Depending on the analysed locus (promoter, gene body) we found region-dependent significant LAG3 methylation differences between monocytes, B cells, CD8+ and CD4+ T cells, regulatory T cells, melanocytes, and melanoma cell lines. In tumor tissues, methylation correlated significantly with LAG3 mRNA expression, immune cell infiltrates (histopathologic lymphocyte score and RNA-Seq signatures of distinct immune infiltrates), and an interferon-γ signature. Finally, LAG3 methylation was associated with overall survival in the TCGA cohort and progression-free survival in the ICB cohort. We detected basal LAG3 mRNA expression in the melanoma cell A375 and an interferon-γ inducible expression after demethylation with 5-azacytidine. INTERPRETATION: Our study points towards an epigenetic regulation of LAG3 via promoter methylation and suggests a prognostic and predictive significance of LAG3 methylation in melanoma. Our results give insight in the tumor cell-intrinsic transcriptional regulation of LAG3 in melanoma. In perspective, our results might pave the way for investigating LAG3 methylation as a predictive biomarker for response to anti-LAG3 immune checkpoint blockage. FUNDING: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.


Asunto(s)
Antígenos CD/genética , Biomarcadores de Tumor , Metilación de ADN , Melanoma/etiología , Melanoma/patología , Regiones Promotoras Genéticas , Línea Celular Tumoral , Epigénesis Genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Melanoma/mortalidad , Melanoma/terapia , Terapia Molecular Dirigida , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Resultado del Tratamiento , Microambiente Tumoral , Proteína del Gen 3 de Activación de Linfocitos
11.
J Immunother Cancer ; 7(1): 50, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30786924

RESUMEN

BACKGROUND: Long-term survival of stage IV melanoma patients has improved significantly with the development of immune checkpoint inhibitors (CIs). Reliable biomarkers to predict response and clinical outcome are needed. METHODS: We investigated the role of melanoma-associated antibodies as predictive markers for CI therapy in two independent cohorts. In cohort 1, a prospective study, we measured specific antibodies before treatment, after one week and after six to nine weeks of treatment. Cohort 2 consisted of serum samples prior to CI therapy initiation. ELISA assays were performed to quantify specific IgG directed against melanocyte differentiation antigens tyrosinase-related proteins 1 and 2 (TRP1/TYRP1 and TRP2/TYRP2), glycoprotein 100 (gp100), MelanA/MART1, and the cancer-testis antigen NY-ESO-1. Response was defined as either complete or partial remission on CT scan according to RECIST 1.1. RESULTS: In cohort 1, baseline levels of these antibodies were higher in the responder group, although statistical significance was only reached for NY-ESO-1 (p = 0.007). In cohort 2, significantly higher antibody baseline levels for MelanA/MART1 (p = 0.003) and gp100 (p = 0.029) were found. After pooling the results from both cohorts, higher levels of MelanA/MART1 (p = 0.013), TRP1/TYRP1 (p = 0.048), TRP2/TYRP2 (p = 0.047) and NY-ESO-1 (p = 0.005) specific antibodies at baseline were independently associated with response. CONCLUSIONS: Melanoma-associated antibodies may be candidate biomarkers for response and survival in metastatic melanoma patients being treated with CIs. These markers may be used to complement patient assessment, in combination with PD-L1 status, tumor-infiltrating lymphocytes and tumor mutational burden, with the aim to predict outcome of CI treatment in patients with metastatic melanoma. TRIAL REGISTRATION: Ethikkommission Ostschweiz, EKOS 16/079 https://ongoingprojects.swissethics.ch/runningProjects_list.php?q=%28BASECID~contains~2016-00998%29&orderby=dBASECID .


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Antineoplásicos/sangre , Antineoplásicos Inmunológicos/uso terapéutico , Ipilimumab/uso terapéutico , Melanoma/sangre , Nivolumab/uso terapéutico , Anciano , Anciano de 80 o más Años , Biomarcadores , Femenino , Humanos , Inmunoglobulina G/sangre , Masculino , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Persona de Mediana Edad
12.
JAMA Oncol ; 5(7): 1043-1047, 2019 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-31021392

RESUMEN

IMPORTANCE: Immunotherapy with checkpoint inhibitors targeting the PD-1 (programmed cell death 1) axis has brought notable progress in patients with non-small cell lung cancer (NSCLC) and other cancers. However, autoimmune toxic effects are frequent and poorly understood, making it important to understand the pathophysiologic processes of autoimmune adverse effects induced by checkpoint inhibitor therapy. OBJECTIVE: To gain mechanistic insight into autoimmune skin toxic effects induced by anti-PD-1 treatment in patients with non-small cell lung cancer. DESIGN, SETTING, AND PARTICIPANTS: This prospective cohort study was conducted from July 1, 2016, to December 31, 2018. Patients (n = 73) with non-small cell lung cancer who received anti-PD-1 therapy (nivolumab or pembrolizumab) were recruited from 4 different centers in Switzerland (Kantonsspital St Gallen, Spital Grabs, Spital Wil, and Spital Flawil). Peripheral blood mononuclear cells, tumor biopsy specimens and biopsies from sites of autoimmune skin toxic effects were collected over a 2-year period, with patient follow-up after 1 year. MAIN OUTCOMES AND MEASURES: Response to treatment, overall survival, progression-free survival, and development of autoimmune toxic effects (based on standard laboratory values and clinical examinations). RESULTS: Of the cohort of 73 patients with NSCLC (mean [SD] age, 68.1 [8.9] years; 44 [60%] men), 25 (34.2% [95% CI, 24.4%-45.7%]) developed autoimmune skin toxic effects, which were more frequent in patients with complete remission or partial remission (68.2% [95% CI, 47.3%-83.6%]) than those with progressive or stable disease (19.6% [95% CI, 11.0%-32.5%]) (χ2 = 14.02, P < .001). Nine T-cell antigens shared between tumor tissue and skin were identified. These antigens were able to stimulate CD8+ and CD4+ T cells in vitro. Several of the antigen-specific T cells found in blood samples were also present in autoimmune skin lesions and lung tumors of patients who responded to anti-PD-1 therapy. CONCLUSIONS AND RELEVANCE: These findings highlight a potential mechanism of checkpoint inhibitor-mediated autoimmune toxic effects and describe the association between toxic effects and response to therapy; such an understanding will help in controlling adverse effects, deciphering new cancer antigens, and further improving immunotherapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados/efectos adversos , Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/efectos adversos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Nivolumab/efectos adversos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/inmunología , Anciano , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
13.
Methods Mol Biol ; 1404: 351-364, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27076310

RESUMEN

Vaccination with a recombinant LCMV based vector expressing tumor-associated or viral antigens is a safe and versatile method to induce an immune response against tumors or viral infections. Here, we describe the generation of recombinant LCMV vectors in which the gene encoding the viral LCMV-GP was substituted with a gene of interest (vaccine antigen). This renders the vaccine vector propagation-incompetent while it preserves the property of eliciting a strong cytotoxic T cell response.


Asunto(s)
Ingeniería Genética/métodos , Vectores Genéticos/genética , Virus de la Coriomeningitis Linfocítica/genética , Vacunas/genética , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Clonación Molecular , ADN Recombinante/genética , Inmunogenicidad Vacunal , Ratones , Plásmidos/genética , Bazo/inmunología , Coloración y Etiquetado , Transfección , Vacunas/inmunología
14.
Fam Syst Health ; 33(1): 61-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25581558

RESUMEN

Prolonged, intensive treatment protocols for childhood cancer disrupt family routines and daily functioning, with effects extending to all family members. Despite their unique needs, siblings of children with cancer receive limited attention from community organizations and researchers. Community-academic partnerships may foster research that effectively assesses and addresses siblings' unmet needs. In this article, "community" refers to siblings of children with cancer who participate in SuperSibs!, a national nonprofit organization for siblings of children with cancer. This article (a) describes a replicable model for successful community-academic partnerships: the Sibling Research Advisory Board (SRAB) and (b) articulates "lessons learned" from this partnership, including documenting the ability to recruit a representative sample through a community organization. Lessons emerged from an iterative process of discussion and revision that involved all SRAB members. This case study describes approaches to overcoming practical obstacles in community-partnered research planning and implementation. To meet the common goals of identifying and addressing unmet sibling needs, SRAB partners learned to establish a common language, identify each team member's unique expertise, and acknowledge differences in approach (e.g., methodology, pace of accomplishment) between research and community service. SRAB's ability to recruit a representative sample was achieved through close collaboration with SuperSibs! and implementation of active recruitment strategies to overcome barriers to research participation. Protection of community member privacy was emphasized alongside methodological rigor. Community-academic partnerships enable research with high-need, hard-to-access populations. Proactively identifying and addressing common pitfalls of community-academic partnerships promotes community engagement and acceptability and facilitates high-quality research.


Asunto(s)
Neoplasias/psicología , Relaciones entre Hermanos , Adolescente , Niño , Participación de la Comunidad , Investigación Participativa Basada en la Comunidad , Familia , Humanos , Neoplasias/complicaciones , Neoplasias/terapia , Organizaciones sin Fines de Lucro/organización & administración , Ajuste Social
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA