Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunity ; 57(2): 223-244, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38354702

RESUMEN

Immune responses must be tightly regulated to ensure both optimal protective immunity and tolerance. Costimulatory pathways within the B7:CD28 family provide essential signals for optimal T cell activation and clonal expansion. They provide crucial inhibitory signals that maintain immune homeostasis, control resolution of inflammation, regulate host defense, and promote tolerance to prevent autoimmunity. Tumors and chronic pathogens can exploit these pathways to evade eradication by the immune system. Advances in understanding B7:CD28 pathways have ushered in a new era of immunotherapy with effective drugs to treat cancer, autoimmune diseases, infectious diseases, and transplant rejection. Here, we discuss current understanding of the mechanisms underlying the coinhibitory functions of CTLA-4, PD-1, PD-L1:B7-1 and PD-L2:RGMb interactions and less studied B7 family members, including HHLA2, VISTA, BTNL2, and BTN3A1, as well as their overlapping and unique roles in regulating immune responses, and the therapeutic potential of these insights.


Asunto(s)
Enfermedades Autoinmunes , Antígenos CD28 , Humanos , Antígenos CD28/metabolismo , Amigos , Linfocitos T , Antígeno CTLA-4/metabolismo , Inmunoterapia , Antígeno B7-1/metabolismo , Inmunoglobulinas/metabolismo , Butirofilinas/metabolismo , Antígenos CD/metabolismo
2.
Clin Cancer Res ; 30(4): 803-813, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38060202

RESUMEN

PURPOSE: Programmed cell death protein 1 (PD-1) expression on CD8+TIM-3-LAG-3- tumor-infiltrating cells predicts positive response to PD-1 blockade in metastatic clear-cell renal cell carcinoma (mccRCC). Because inhibition of PD-1 signaling in regulatory T cells (Treg) augments their immunosuppressive function, we hypothesized that PD-1 expression on tumor-infiltrating Tregs would predict resistance to PD-1 inhibitors. EXPERIMENTAL DESIGN: PD-1+ Tregs were phenotyped using multiparametric immunofluorescence in ccRCC tissues from the CheckMate-025 trial (nivolumab: n = 91; everolimus: n = 90). Expression of CD8, PD-1, TIM-3, and LAG-3 was previously determined (Ficial and colleagues, 2021). Clinical endpoints included progression-free survival (PFS), overall survival (OS), and objective response rate (ORR). RESULTS: In the nivolumab (but not everolimus) arm, high percentage of PD-1+ Tregs was associated with shorter PFS (3.19 vs. 5.78 months; P = 0.021), shorter OS (18.1 vs. 27.7 months; P = 0.013) and marginally lower ORR (12.5% vs. 31.3%; P = 0.059). An integrated biomarker (PD-1 Treg/CD8 ratio) was developed by calculating the ratio between percentage of PD-1+Tregs (marker of resistance) and percentage of CD8+PD-1+TIM-3-LAG-3- cells (marker of response). In the nivolumab (but not everolimus) arm, patients with high PD-1 Treg/CD8 ratio experienced shorter PFS (3.48 vs. 9.23 months; P < 0.001), shorter OS (18.14 vs. 38.21 months; P < 0.001), and lower ORR (15.69% vs. 40.00%; P = 0.009). Compared with the individual biomarkers, the PD-1 Treg/CD8 ratio showed improved ability to predict outcomes to nivolumab versus everolimus. CONCLUSIONS: PD-1 expression on Tregs is associated with resistance to PD-1 blockade in mccRCC, suggesting that targeting Tregs may synergize with PD-1 inhibition. A model that integrates PD-1 expression on Tregs and CD8+TIM-3-LAG-3- cells has higher predictive value.


Asunto(s)
Carcinoma de Células Renales , Humanos , Carcinoma de Células Renales/patología , Nivolumab/uso terapéutico , Linfocitos T Reguladores/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Everolimus/uso terapéutico , Receptor de Muerte Celular Programada 1/metabolismo
3.
Trends Immunol ; 44(5): 326-328, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37031062

RESUMEN

In a recent article, Puig-Saus et al. computationally predict and experimentally validate neoantigen-specific T cell responses in patients with melanoma. They identify a restricted set of neoantigens recognized by polyclonal CD8+ T cells as a unique feature of anti-PD-1 responders and engineer autologous tumor-responsive T cells expressing neoantigen-specific TCRs, providing proof-of-concept for future cellular therapies.


Asunto(s)
Linfocitos T CD8-positivos , Melanoma , Humanos , Antígenos de Neoplasias , Melanoma/terapia , Receptores de Antígenos de Linfocitos T
4.
Curr Opin Immunol ; 80: 102283, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36709596

RESUMEN

Immune checkpoint receptors such as programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte associated protein 4 (CTLA-4), lymphocyte-activation gene 3 (LAG-3), and T cell immunoglobulin and ITIM domain (TIGIT) have distinct and overlapping inhibitory functions that regulate Tcell activation, differentiation, and function. These inhibitory receptors also mediate tolerance, and dysregulation of these receptors can result in a breach of tolerance and the development of autoimmune syndromes. Similarly, antibody blockade of immune checkpoint receptors or their ligands for cancer immunotherapy may trigger a spectrum of organ inflammation that resembles autoimmunity, termed immune-related adverse events (irAE). In this review, we discuss recent advances in the regulation of autoimmunity by immune checkpoint receptors. We highlight coordinated gene expression programs linking checkpoint receptors, heterogeneity within autoreactive T-cell populations, parallels between irAE and autoimmunity, and bidirectional functional interactions between immune checkpoint receptors and their ligands.


Asunto(s)
Enfermedades del Sistema Inmune , Neoplasias , Humanos , Autoinmunidad , Ligandos , Antígeno CTLA-4 , Inmunoterapia , Linfocitos T , Receptores Inmunológicos/metabolismo , Enfermedades del Sistema Inmune/metabolismo
5.
Sci Adv ; 8(15): eabj3286, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35417234

RESUMEN

Trogocytosis modulates immune responses, with still unclear underlying molecular mechanisms. Using leukemia mouse models, we found that lymphocytes perform trogocytosis at high rates with tumor cells. While performing trogocytosis, both Natural Killer (NK) and CD8+ T cells acquire the checkpoint receptor PD-1 from leukemia cells. In vitro and in vivo investigation revealed that PD-1 on the surface of NK cells, rather than being endogenously expressed, was derived entirely from leukemia cells in a SLAM receptor-dependent fashion. PD-1 acquired via trogocytosis actively suppressed NK cell antitumor immunity. PD-1 trogocytosis was corroborated in patients with clonal plasma cell disorders, where NK cells that stained for PD-1 also stained for tumor cell markers. Our results, in addition to shedding light on a previously unappreciated mechanism underlying the presence of PD-1 on NK and cytotoxic T cells, reveal the immunoregulatory effect of membrane transfer occurring when immune cells contact tumor cells.


Asunto(s)
Leucemia , Neoplasias , Animales , Linfocitos T CD8-positivos , Humanos , Células Asesinas Naturales , Leucemia/metabolismo , Ratones , Neoplasias/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo
6.
Nat Med ; 27(3): 426-433, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33664492

RESUMEN

Metastatic castration-resistant prostate cancer is typically lethal, exhibiting intrinsic or acquired resistance to second-generation androgen-targeting therapies and minimal response to immune checkpoint inhibitors1. Cellular programs driving resistance in both cancer and immune cells remain poorly understood. We present single-cell transcriptomes from 14 patients with advanced prostate cancer, spanning all common metastatic sites. Irrespective of treatment exposure, adenocarcinoma cells pervasively coexpressed multiple androgen receptor isoforms, including truncated isoforms hypothesized to mediate resistance to androgen-targeting therapies2,3. Resistance to enzalutamide was associated with cancer cell-intrinsic epithelial-mesenchymal transition and transforming growth factor-ß signaling. Small cell carcinoma cells exhibited divergent expression programs driven by transcriptional regulators promoting lineage plasticity and HOXB5, HOXB6 and NR1D2 (refs. 4-6). Additionally, a subset of patients had high expression of dysfunction markers on cytotoxic CD8+ T cells undergoing clonal expansion following enzalutamide treatment. Collectively, the transcriptional characterization of cancer and immune cells from human metastatic castration-resistant prostate cancer provides a basis for the development of therapeutic approaches complementing androgen signaling inhibition.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Próstata Resistentes a la Castración/terapia , Transcripción Genética/efectos de los fármacos , Biopsia , Linfocitos T CD8-positivos/inmunología , Resistencia a Antineoplásicos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/metabolismo
7.
Cancer Cell ; 39(5): 632-648.e8, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33711273

RESUMEN

The tumor immune microenvironment plays a critical role in cancer progression and response to immunotherapy in clear cell renal cell carcinoma (ccRCC), yet the composition and phenotypic states of immune cells in this tumor are incompletely characterized. We performed single-cell RNA and T cell receptor sequencing on 164,722 individual cells from tumor and adjacent non-tumor tissue in patients with ccRCC across disease stages: early, locally advanced, and advanced/metastatic. Terminally exhausted CD8+ T cells were enriched in metastatic disease and were restricted in T cell receptor diversity. Within the myeloid compartment, pro-inflammatory macrophages were decreased, and suppressive M2-like macrophages were increased in advanced disease. Terminally exhausted CD8+ T cells and M2-like macrophages co-occurred in advanced disease and expressed ligands and receptors that support T cell dysfunction and M2-like polarization. This immune dysfunction circuit is associated with a worse prognosis in external cohorts and identifies potentially targetable immune inhibitory pathways in ccRCC.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma de Células Renales/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Renales/genética , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunoterapia/métodos , Neoplasias Renales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Macrófagos/metabolismo , Microambiente Tumoral/inmunología
8.
J Exp Med ; 218(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33651880

RESUMEN

The ability to monitor anti-tumor CD8+ T cell responses in the blood has tremendous therapeutic potential. Here, we used paired single-cell RNA and TCR sequencing to detect and characterize "tumor-matching" (TM) CD8+ T cells in the blood of mice with MC38 tumors or melanoma patients using the TCR as a molecular barcode. TM cells showed increased activation compared with nonmatching T cells in blood and were less exhausted than matching cells in tumors. Importantly, PD-1, which has been used to identify putative circulating anti-tumor CD8+ T cells, showed poor sensitivity for identifying TM cells. By leveraging the transcriptome, we identified candidate cell surface markers for TM cells in mice and patients and validated NKG2D, CD39, and CX3CR1 in mice. These data show that the TCR can be used to identify tumor-relevant cells for characterization, reveal unique transcriptional properties of TM cells, and develop marker panels for tracking and analysis of these cells.


Asunto(s)
Adenocarcinoma/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias del Colon/inmunología , Melanoma/sangre , Melanoma/inmunología , Análisis de la Célula Individual/métodos , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/inmunología , Adenocarcinoma/patología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/patología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transcriptoma
9.
J Exp Med ; 218(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33601411

RESUMEN

The treatment of many cancers has been revolutionized by immune checkpoint blockade (ICB) as a standard-of-care therapeutic. Despite many successes, a large proportion of patients treated with ICB agents experience immune-related adverse events (irAEs) in the form of clinical autoimmunity, ranging from mild to life threatening, that can limit cancer treatment. A mechanistic understanding of these irAEs is required to better treat or prevent irAEs and to predict those patients who are susceptible to irAEs. We propose several mechanisms that may contribute to the generation of irAEs: (1) preexisting susceptibility to autoimmunity, (2) aberrant presentation of "self" by the tumor, and (3) loss of tolerance driven by the tumor or tissue microenvironment.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico , Tolerancia Inmunológica/efectos de los fármacos , Inmunoterapia/efectos adversos , Neoplasias , Microambiente Tumoral , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
10.
Cell Rep ; 31(13): 107827, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32610128

RESUMEN

The PD-1 pathway regulates dysfunctional T cells in chronic infection and cancer, but the role of this pathway during acute infection remains less clear. Here, we demonstrate that PD-1 signals are needed for optimal memory. Mice deficient in the PD-1 pathway exhibit impaired CD8+ T cell memory following acute influenza infection, including reduced virus-specific CD8+ T cell numbers and compromised recall responses. PD-1 blockade during priming leads to similar differences early post-infection but without the defect in memory formation, suggesting that timing and/or duration of PD-1 blockade could be tailored to modulate host responses. Our studies reveal a role for PD-1 as an integrator of CD8+ T cell signals that promotes CD8+ T cell memory formation and suggest PD-1 continues to fine-tune CD8+ T cells after they migrate into non-lymphoid tissues. These findings have important implications for PD-1-based immunotherapy, in which PD-1 inhibition may influence memory responses in patients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Subtipo H3N2 del Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Receptor de Muerte Celular Programada 1/metabolismo , Transducción de Señal , Administración Intranasal , Animales , Muerte Celular/inmunología , Diferenciación Celular/inmunología , Proliferación Celular , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/patología , Especificidad de la Especie
11.
Nat Med ; 25(3): 389-402, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30842677

RESUMEN

Despite impressive durable responses, immune checkpoint inhibitors do not provide a long-term benefit to the majority of patients with cancer. Understanding genomic correlates of response and resistance to checkpoint blockade may enhance benefits for patients with cancer by elucidating biomarkers for patient stratification and resistance mechanisms for therapeutic targeting. Here we review emerging genomic markers of checkpoint blockade response, including those related to neoantigens, antigen presentation, DNA repair, and oncogenic pathways. Compelling evidence also points to a role for T cell functionality, checkpoint regulators, chromatin modifiers, and copy-number alterations in mediating selective response to immune checkpoint blockade. Ultimately, efforts to contextualize genomic correlates of response into the larger understanding of tumor immune biology will build a foundation for the development of novel biomarkers and therapies to overcome resistance to checkpoint blockade.


Asunto(s)
Presentación de Antígeno/genética , Antígenos de Neoplasias/genética , Antineoplásicos Inmunológicos/uso terapéutico , Reparación del ADN/genética , Genoma/genética , Neoplasias/tratamiento farmacológico , Linfocitos T/inmunología , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Antígeno B7-H1/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Carcinogénesis/genética , Ensamble y Desensamble de Cromatina/genética , Variaciones en el Número de Copia de ADN/genética , Resistencia a Antineoplásicos/genética , Humanos , Mutación , Neoplasias/genética , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal
13.
J Clin Invest ; 128(10): 4441-4453, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30179225

RESUMEN

BACKGROUND: Understanding the integrated immunogenomic landscape of advanced prostate cancer (APC) could impact stratified treatment selection. METHODS: Defective mismatch repair (dMMR) status was determined by either loss of mismatch repair protein expression on IHC or microsatellite instability (MSI) by PCR in 127 APC biopsies from 124 patients (Royal Marsden [RMH] cohort); MSI by targeted panel next-generation sequencing (MSINGS) was then evaluated in the same cohort and in 254 APC samples from the Stand Up To Cancer/Prostate Cancer Foundation (SU2C/PCF). Whole exome sequencing (WES) data from this latter cohort were analyzed for pathogenic MMR gene variants, mutational load, and mutational signatures. Transcriptomic data, available for 168 samples, was also performed. RESULTS: Overall, 8.1% of patients in the RMH cohort had some evidence of dMMR, which associated with decreased overall survival. Higher MSINGS scores associated with dMMR, and these APCs were enriched for higher T cell infiltration and PD-L1 protein expression. Exome MSINGS scores strongly correlated with targeted panel MSINGS scores (r = 0.73, P < 0.0001), and higher MSINGS scores associated with dMMR mutational signatures in APC exomes. dMMR mutational signatures also associated with MMR gene mutations and increased immune cell, immune checkpoint, and T cell-associated transcripts. APC with dMMR mutational signatures overexpressed a variety of immune transcripts, including CD200R1, BTLA, PD-L1, PD-L2, ADORA2A, PIK3CG, and TIGIT. CONCLUSION: These data could impact immune target selection, combination therapeutic strategy selection, and selection of predictive biomarkers for immunotherapy in APC. FUNDING: We acknowledge funding support from Movember, Prostate Cancer UK, The Prostate Cancer Foundation, SU2C, and Cancer Research UK.


Asunto(s)
Antígeno B7-H1 , Reparación de la Incompatibilidad de ADN , Inmunoterapia , Inestabilidad de Microsatélites , Mutación , Proteínas de Neoplasias , Neoplasias de la Próstata , Adulto , Anciano , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia
14.
Cancer Discov ; 8(9): 1096-1111, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29903880

RESUMEN

Clinically relevant subtypes exist for pancreatic ductal adenocarcinoma (PDAC), but molecular characterization is not yet standard in clinical care. We implemented a biopsy protocol to perform time-sensitive whole-exome sequencing and RNA sequencing for patients with advanced PDAC. Therapeutically relevant genomic alterations were identified in 48% (34/71) and pathogenic/likely pathogenic germline alterations in 18% (13/71) of patients. Overall, 30% (21/71) of enrolled patients experienced a change in clinical management as a result of genomic data. Twenty-six patients had germline and/or somatic alterations in DNA-damage repair genes, and 5 additional patients had mutational signatures of homologous recombination deficiency but no identified causal genomic alteration. Two patients had oncogenic in-frame BRAF deletions, and we report the first clinical evidence that this alteration confers sensitivity to MAPK pathway inhibition. Moreover, we identified tumor/stroma gene expression signatures with clinical relevance. Collectively, these data demonstrate the feasibility and value of real-time genomic characterization of advanced PDAC.Significance: Molecular analyses of metastatic PDAC tumors are challenging due to the heterogeneous cellular composition of biopsy specimens and rapid progression of the disease. Using an integrated multidisciplinary biopsy program, we demonstrate that real-time genomic characterization of advanced PDAC can identify clinically relevant alterations that inform management of this difficult disease. Cancer Discov; 8(9); 1096-111. ©2018 AACR.See related commentary by Collisson, p. 1062This article is highlighted in the In This Issue feature, p. 1047.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Perfilación de la Expresión Génica/métodos , Variación Genética , Genómica/métodos , Neoplasias Pancreáticas/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/tratamiento farmacológico , Reparación del ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Mutación de Línea Germinal , Recombinación Homóloga , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/tratamiento farmacológico , Medicina de Precisión , Análisis de Secuencia de ARN/métodos , Secuenciación del Exoma/métodos
15.
Clin Cancer Res ; 22(23): 5642-5650, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27698000

RESUMEN

Immunotherapy has led to a paradigm shift in the treatment of some malignancies, providing long-term, durable responses for patients with advanced cancers. However, such therapy has benefited only a subset of patients, with some patients failing to respond to treatment at all and others achieving a limited response followed by tumor progression. Understanding factors contributing to an effective response and further elucidating mechanisms of resistance will be crucial as these therapies are applied more broadly. Genomics-based approaches have significantly advanced the study of response and resistance to immunotherapy in general, and to immune checkpoint blockade more specifically. Here, we review how genomic and transcriptomic approaches have identified both somatic and germline positive correlates of response, including high mutational/neoantigen load and low intratumoral heterogeneity, among others. The genomic analysis of resistant tumors has additionally identified crucial factors involved in resistance to immune checkpoint blockade, including loss of PTEN and upregulation of other immune checkpoints. Overall, the continued use of genomic techniques at the point of care, combined with appropriate functional studies, would ideally lead to a better understanding of why certain patients respond to immune-based therapies, allowing clinicians to identify the subset of patients likely to benefit from such therapy, and potentially providing insight into how other therapies may be added in combination to increase the number of patients who may benefit from immunotherapy. Clin Cancer Res; 22(23); 5642-50. ©2016 AACR.


Asunto(s)
Resistencia a Antineoplásicos/genética , Neoplasias/genética , Transcriptoma/genética , Animales , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/inmunología , Genómica/métodos , Humanos , Inmunoterapia/métodos , Mutación/efectos de los fármacos , Mutación/genética , Neoplasias/inmunología , Neoplasias/terapia , Transcriptoma/efectos de los fármacos , Transcriptoma/inmunología
16.
J Immunol ; 188(10): 5177-88, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22508927

RESUMEN

Vaccines designed to prevent or to treat hepatitis C viral infection must achieve maximum cross-reactivity against widely divergent circulating strains. Rational approaches for sequence selection to maximize immunogenicity and minimize genetic distance across circulating strains may enhance vaccine induction of optimal cytotoxic T cell responses. We assessed T cell recognition of potential hepatitis C virus (HCV) vaccine sequences generated using three rational approaches: combining epitopes with predicted tight binding to the MHC, consensus sequence (most common amino acid at each position), and representative ancestral sequence that had been derived using bayesian phylogenetic tools. No correlation was seen between peptide-MHC binding affinity and frequency of recognition, as measured by an IFN-γ T cell response in HLA-matched HCV-infected individuals. Peptides encoding representative, consensus, and natural variant sequences were then tested for the capacity to expand CD8 T cell populations and to elicit cross-reactive CD8 T cell responses. CD8(+) T cells expanded with representative sequence HCV generally more broadly and robustly recognized highly diverse circulating HCV strains than did T cells expanded with either consensus sequence or naturally occurring sequence variants. These data support the use of representative sequence in HCV vaccine design.


Asunto(s)
Hepacivirus/inmunología , Hepatitis C/inmunología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Línea Celular , Células Cultivadas , Estudios de Cohortes , Secuencia de Consenso/inmunología , Reacciones Cruzadas/inmunología , Antígenos HLA/inmunología , Hepatitis C/metabolismo , Hepatitis C/virología , Humanos , Fragmentos de Péptidos/síntesis química , Estudios Prospectivos , Vacunas contra Hepatitis Viral/síntesis química , Vacunas contra Hepatitis Viral/inmunología , Vacunas contra Hepatitis Viral/metabolismo
17.
J Virol ; 86(10): 5915-21, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22438535

RESUMEN

Hepatitis C virus (HCV) research is hampered by the use of arbitrary representative isolates in cell culture and immunology. The most replicative isolate in vitro is a subtype 2a virus (JFH-1); however, genotype 1 is more prevalent worldwide and represents about 70% of infections in the United States, and genotypes differ from one another by 31% to 33% at the nucleotide level. For phylogenetic and immunologic analyses, viruses H77 and HCV-1 (both subtype 1a) are commonly used based on their historic importance. In an effort to rationally design a representative subtype 1a virus (Bole1a), we used Bayesian phylogenetics, ancestral sequence reconstruction, and covariance analysis on a curated set of 390 full-length human HCV 1a sequences from GenBank. By design, Bole1a contains variations present in widely circulating strains and matches more epitope-sized peptides in a full-genome comparison to subtype 1a isolates than any other sequence studied. Parallel analyses confirm that selected epitopes from the Bole1a genome were able to elicit a robust T cell response. In a proof of concept for infectivity, the envelope genes (E1 and E2) of Bole1a were expressed in an HIV pseudoparticle system containing HCV envelope genes and HIV nonenvelope genes with luciferase expression. The resulting Bole1a pseudoparticle robustly infected Hep3B cells. In this study, we demonstrate that a rationally designed, fully synthetic HCV genome contains representative epitopes and envelope genes that assemble properly and mediate entry into target cells.


Asunto(s)
Genes Sintéticos , Genoma Viral , Hepacivirus/genética , Hepatitis C/virología , Línea Celular , Biología Computacional , Genotipo , Hepacivirus/química , Hepacivirus/clasificación , Hepacivirus/fisiología , Humanos , Datos de Secuencia Molecular , Filogenia , Proteínas Virales/síntesis química , Proteínas Virales/genética , Proteínas Virales/metabolismo , Internalización del Virus
18.
Immunol Res ; 47(1-3): 216-27, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20066508

RESUMEN

Hepatitis C virus (HCV) infects over 170 million people worldwide and is a leading cause of cirrhosis and hepatocellular carcinoma. Approximately 20% [corrected] of those acutely infected clear the infection, whereas the remaining 80% [corrected] progress to chronic infection. Hepatitis C thus provides a model in which successful and unsuccessful responses can be compared to better understand the human response to viral infection. Our laboratory studies the strategies by which HCV evades the adaptive immune response. This review describes the impact of viral mutation on T cell recognition, the role of cell surface inhibitory receptors in recognition of HCV, and the development of antibodies that neutralize HCV infection. Understanding what constitutes an effective immune response in the control of HCV may enable the development of prophylactic and therapeutic vaccines for HCV and other chronic viral infections.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Hepacivirus/fisiología , Hepatitis C Crónica/inmunología , Evasión Inmune , Linfocitos T/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Epítopos de Linfocito T/genética , Hepacivirus/genética , Hepacivirus/inmunología , Hepatitis C Crónica/virología , Humanos , Mutación , Linfocitos T/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...