Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38683966

RESUMEN

Relapse is the leading cause of death after allogeneic hematopoietic stem cell transplantation (HCT) for leukemia. T cells engineered by gene transfer to express T cell receptors (TCR; TCR-T) specific for hematopoietic-restricted minor histocompatibility (H) antigens may provide a potent selective anti-leukemic effect post-HCT. We conducted a phase I clinical trial employing a novel TCR-T product targeting the minor H antigen HA-1 to treat or consolidate treatment of persistent or recurrent leukemia and myeloid neoplasms. The primary objective was to evaluate the feasibility and safety of administration of HA-1 TCR-T post-HCT. CD8+ and CD4+ T cells expressing the HA-1 TCR and a CD8-co-receptor were successfully manufactured from HA-1 disparate HCT donors. One or more infusions of HA-1 TCR-T following lymphodepleting chemotherapy were administered to nine HCT recipients who had developed disease recurrence post-HCT. TCR-T cells expanded and persisted in vivo after adoptive transfer. No dose-limiting toxicities occurred. Although the study was not designed to assess efficacy, four patients achieved or maintained complete remissions following lymphodepletion and HA-1 TCR-T, with one ongoing at >2 years. Single-cell RNA sequencing of relapsing/progressive leukemia after TCR-T therapy identified upregulated molecules associated with T cell dysfunction or cancer cell survival. HA-1 TCR-T therapy appears feasible and safe and shows preliminary signals of efficacy. This clinical trial is registered at clinicaltrials.gov as NCT03326921.

2.
Immunity ; 45(2): 389-401, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27521269

RESUMEN

CD8(+) T cells recognizing tumor-specific antigens are detected in cancer patients but are dysfunctional. Here we developed a tamoxifen-inducible liver cancer mouse model with a defined oncogenic driver antigen (SV40 large T-antigen) to follow the activation and differentiation of naive tumor-specific CD8(+) T (TST) cells after tumor initiation. Early during the pre-malignant phase of tumorigenesis, TST cells became dysfunctional, exhibiting phenotypic, functional, and transcriptional features similar to dysfunctional T cells isolated from late-stage human tumors. Thus, T cell dysfunction seen in advanced human cancers may already be established early during tumorigenesis. Although the TST cell dysfunctional state was initially therapeutically reversible, it ultimately evolved into a fixed state. Persistent antigen exposure rather than factors associated with the tumor microenvironment drove dysfunction. Moreover, the TST cell differentiation and dysfunction program exhibited features distinct from T cell exhaustion in chronic infections. Strategies to overcome this antigen-driven, cell-intrinsic dysfunction may be required to improve cancer immunotherapy.


Asunto(s)
Antígenos Transformadores de Poliomavirus/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias Hepáticas/inmunología , Animales , Carcinogénesis , Diferenciación Celular , Células Cultivadas , Senescencia Celular , Modelos Animales de Enfermedad , Humanos , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/terapia , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tamoxifeno , Microambiente Tumoral
3.
Gynecol Oncol ; 160(2): 427-437, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33229044

RESUMEN

OBJECTIVES: Mouse models of ovarian cancer commonly transfer large numbers of tumor cells into the peritoneal cavity to establish experimental metastatic disease, which may not adequately model early metastatic spread from a primary tumor site. We hypothesized we could develop an ovarian cancer model that predictably represents micro-metastatic disease. METHODS: Murine ID8VEGF ovarian cancer cells were transduced to express enhanced luciferase (eLuc) to enable intravital detection of microscopic disease burden and injected beneath the ovarian bursa of C57Bl/6 mice. At 6 or 10 weeks after orthotopic injection, when mice had detectable metastases, hysterectomy and bilateral salpingo-oophorectomy was performed to remove all macroscopic disease, and survival monitored. Immunohistochemistry and gene expression profiling were performed on primary and metastatic tumors. RESULTS: eLuc-transduced ID8VEGF cells were brighter than cells transduced with standard luciferase, enabling in vivo visualization of microscopic intra-abdominal metastases developing after orthotopic injection. Primary surgical cytoreduction removed the primary tumor mass but left minimal residual disease in all mice. Metastatic sites that developed following orthotopic injection were similar to metastatic human ovarian cancer sites. Gene expression and immune infiltration were similar between primary and metastatic mouse tumors. Surgical cytoreduction prolonged survival compared to no surgery, with earlier cytoreduction more beneficial than delayed, despite micro-metastatic disease in both settings. CONCLUSIONS: Mice with primary ovarian tumors established through orthotopic injection develop progressively fatal metastatic ovarian cancer, and benefit from surgical cytoreduction to remove bulky disease. This model enables the analysis of therapeutic regimens designed to target and potentially eradicate established minimal residual disease.


Asunto(s)
Procedimientos Quirúrgicos de Citorreducción , Modelos Animales de Enfermedad , Micrometástasis de Neoplasia/terapia , Neoplasias Ováricas/cirugía , Neoplasias Peritoneales/cirugía , Animales , Línea Celular Tumoral/trasplante , Femenino , Humanos , Histerectomía , Ratones , Neoplasia Residual , Neoplasias Ováricas/patología , Ovario/patología , Ovario/cirugía , Cavidad Peritoneal/patología , Cavidad Peritoneal/cirugía , Neoplasias Peritoneales/secundario , Salpingooforectomía , Carga Tumoral
4.
J Immunol ; 200(3): 1088-1100, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29288199

RESUMEN

Adoptive T cell therapies have achieved significant clinical responses, especially in hematopoietic cancers. Two types of receptor systems have been used to redirect the activity of T cells, normal heterodimeric TCRs or synthetic chimeric Ag receptors (CARs). TCRs recognize peptide-HLA complexes whereas CARs typically use an Ab-derived single-chain fragments variable that recognizes cancer-associated cell-surface Ags. Although both receptors mediate diverse effector functions, a quantitative comparison of the sensitivity and signaling capacity of TCRs and CARs has been limited due to their differences in affinities and ligands. In this study we describe their direct comparison by using TCRs that could be formatted either as conventional αß heterodimers, or as single-chain fragments variable constructs linked to CD3ζ and CD28 signaling domains or to CD3ζ alone. Two high-affinity TCRs (KD values of ∼50 and 250 nM) against MART1/HLA-A2 or WT1/HLA-A2 were used, allowing MART1 or WT1 peptide titrations to easily assess the impact of Ag density. Although CARs were expressed at higher surface levels than TCRs, they were 10-100-fold less sensitive, even in the absence of the CD8 coreceptor. Mathematical modeling demonstrated that lower CAR sensitivity could be attributed to less efficient signaling kinetics. Furthermore, reduced cytokine secretion observed at high Ag density for both TCRs and CARs suggested a role for negative regulators in both systems. Interestingly, at high Ag density, CARs also mediated greater maximal release of some cytokines, such as IL-2 and IL-6. These results have implications for the next-generation design of receptors used in adoptive T cell therapies.


Asunto(s)
Afinidad de Anticuerpos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno MART-1/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas WT1/inmunología , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Antígenos HLA/inmunología , Humanos , Activación de Linfocitos/inmunología , Proteínas Mutantes Quiméricas/inmunología
5.
Blood ; 130(22): 2410-2419, 2017 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-29042364

RESUMEN

Acute myeloid leukemia (AML), the most common adult acute leukemia in the United States, has the poorest survival rate, with 26% of patients surviving 5 years. Adoptive immunotherapy with T cells genetically modified to recognize tumors is a promising and evolving treatment option. However, antitumor activity, particularly in the context of progressive leukemia, can be dampened both by limited costimulation and triggering of immunoregulatory checkpoints that attenuate T-cell responses. Expression of CD200 (OX2), a negative regulator of T-cell function that binds CD200 receptor (CD200R), is commonly increased in leukemia and other malignancies and is associated with poor prognosis in leukemia patients. To appropriate and redirect the inhibitory effects of CD200R signaling on transferred CD8+ T cells, we engineered CD200R immunomodulatory fusion proteins (IFPs) with the cytoplasmic tail replaced by the signaling domain of the costimulatory receptor, CD28. An analysis of a panel of CD200R-CD28 IFP constructs revealed that the most effective costimulation was achieved in IFPs containing a dimerizing motif and a predicted tumor-T-cell distance that facilitates localization to the immunological synapse. T cells transduced with the optimized CD200R-CD28 IFPs exhibited enhanced proliferation and effector function in response to CD200+ leukemic cells in vitro. In adoptive therapy of disseminated leukemia, CD200R-CD28-transduced leukemia-specific CD8 T cells eradicated otherwise lethal disease more efficiently than wild-type cells and bypassed the requirement for interleukin-2 administration to sustain in vivo activity. The transduction of human primary T cells with the equivalent human IFPs increased proliferation and cytokine production in response to CD200+ leukemia cells, supporting clinical translation. This trial was registered at www.clinicaltrials.gov as #NCT01640301.


Asunto(s)
Antígenos de Superficie/genética , Antígenos CD28/genética , Inmunoterapia Adoptiva/métodos , Leucemia/terapia , Glicoproteínas de Membrana/genética , Receptores de Superficie Celular/genética , Linfocitos T/trasplante , Animales , Antígenos de Superficie/inmunología , Antígenos CD28/inmunología , Proliferación Celular , Humanos , Inmunomodulación , Leucemia/genética , Leucemia/inmunología , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Orexina , Receptores de Superficie Celular/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transducción Genética
6.
Proc Natl Acad Sci U S A ; 112(14): E1754-62, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25831525

RESUMEN

Despite the availability of major histocompatibility complex (MHC)-binding peptide prediction algorithms, the development of T-cell vaccines against pathogen and tumor antigens remains challenged by inefficient identification of immunogenic epitopes. CD8(+) T cells must distinguish immunogenic epitopes from nonimmunogenic self peptides to respond effectively against an antigen without endangering the viability of the host. Because this discrimination is fundamental to our understanding of immune recognition and critical for rational vaccine design, we interrogated the biochemical properties of 9,888 MHC class I peptides. We identified a strong bias toward hydrophobic amino acids at T-cell receptor contact residues within immunogenic epitopes of MHC allomorphs, which permitted us to develop and train a hydrophobicity-based artificial neural network (ANN-Hydro) to predict immunogenic epitopes. The immunogenicity model was validated in a blinded in vivo overlapping epitope discovery study of 364 peptides from three HIV-1 Gag protein variants. Applying the ANN-Hydro model on existing peptide-MHC algorithms consistently reduced the number of candidate peptides across multiple antigens and may provide a correlate with immunodominance. Hydrophobicity of TCR contact residues is a hallmark of immunogenic epitopes and marks a step toward eliminating the need for empirical epitope testing for vaccine development.


Asunto(s)
Linfocitos T CD8-positivos/citología , Epítopos de Linfocito T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Adenoviridae/genética , Algoritmos , Aminoácidos/química , Animales , Presentación de Antígeno , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Complejo Mayor de Histocompatibilidad , Ratones , Ratones Endogámicos C57BL , Probabilidad , Unión Proteica , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/química
7.
Immunol Rev ; 257(1): 145-64, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24329795

RESUMEN

Adoptive T-cell therapy involves the isolation, expansion, and reinfusion of T lymphocytes with a defined specificity and function as a means to eradicate cancer. Our research has focused on specifying the requirements for tumor eradication with antigen-specific T cells and T cells transduced to express a defined T-cell receptor (TCR) in mouse models and then translating these strategies to clinical trials. Our design of T-cell-based therapy for cancer has reflected efforts to identify the obstacles that limit sustained effector T-cell activity in mice and humans, design approaches to enhance T-cell persistence, develop methods to increase TCR affinity/T-cell functional avidity, and pursue strategies to overcome tolerance and immunosuppression. With the advent of genetic engineering, a highly functional population of T cells can now be rapidly generated and tailored for the targeted malignancy. Preclinical studies in faithful and informative mouse models, in concert with knowledge gained from analyses of successes and limitations in clinical trials, are shaping how we continue to develop, refine, and broaden the applicability of this approach for cancer therapy.


Asunto(s)
Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Técnicas de Cultivo de Célula , Epítopos de Linfocito T/inmunología , Ingeniería Genética , Terapia Genética , Vectores Genéticos , Humanos , Tolerancia Inmunológica , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Ratones , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Investigación Biomédica Traslacional , Escape del Tumor/inmunología
8.
Trends Immunol ; 35(2): 51-60, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24210163

RESUMEN

CD8 T cell activation and differentiation are tightly controlled, and dependent on the context in which naïve T cells encounter antigen, can either result in functional memory or T cell dysfunction, including exhaustion, tolerance, anergy, or senescence. With the identification of phenotypic and functional traits shared in different settings of T cell dysfunction, distinctions between such dysfunctional states have become blurred. Here, we discuss distinct states of CD8 T cell dysfunction, with an emphasis on: (i) T cell tolerance to self-antigens (self-tolerance); (ii) T cell exhaustion during chronic infections; and (iii) tumor-induced T cell dysfunction. We highlight recent findings on cellular and molecular characteristics defining these states, cell-intrinsic regulatory mechanisms that induce and maintain them, and strategies that can lead to their reversal.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Tolerancia Inmunológica , Animales , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Anergia Clonal , Epigénesis Genética , Humanos , Tolerancia Inmunológica/genética , Memoria Inmunológica , Infecciones/inmunología , Activación de Linfocitos , Ratones , Neoplasias/inmunología , Autotolerancia
9.
Immunity ; 28(5): 662-74, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18424189

RESUMEN

CD8(+) T cell tolerance, although essential for preventing autoimmunity, poses substantial obstacles to eliciting immune responses to tumor antigens, which are generally overexpressed normal proteins. Development of effective strategies to overcome tolerance for clinical applications would benefit from elucidation of the immunologic mechanism(s) regulating T cell tolerance to self. To examine how tolerance is maintained in vivo, we engineered dual-T cell receptor (TCR) transgenic mice in which CD8(+) T cells recognize two distinct antigens: a foreign viral-protein and a tolerizing self-tumor protein. Encounter with peripheral self-antigen rendered dual-TCR T cells tolerant to self, but these cells responded normally through the virus-specific TCR. Moreover, proliferation induced by virus rescued function of tolerized self-tumor-reactive TCR, restoring anti-tumor activity. These studies demonstrate that peripheral CD8(+) T cell tolerance to self-proteins can be regulated at the level of the self-reactive TCR complex rather than by central cellular inactivation and suggest an alternate strategy to enhance adoptive T cell immunotherapy.


Asunto(s)
Autoantígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/inmunología , Autotolerancia/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Ratones , Ratones Mutantes , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal
10.
PLoS Pathog ; 10(9): e1004403, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25254371

RESUMEN

The process of antibody ontogeny typically improves affinity, on-rate, and thermostability, narrows polyspecificity, and rigidifies the combining site to the conformer optimal for binding from the broader ensemble accessible to the precursor. However, many broadly-neutralizing anti-HIV antibodies incorporate unusual structural elements and recognition specificities or properties that often lead to autoreactivity. The ontogeny of 4E10, an autoreactive antibody with unexpected combining site flexibility, was delineated through structural and biophysical comparisons of the mature antibody with multiple potential precursors. 4E10 gained affinity primarily by off-rate enhancement through a small number of mutations to a highly conserved recognition surface. Controverting the conventional paradigm, the combining site gained flexibility and autoreactivity during ontogeny, while losing thermostability, though polyspecificity was unaffected. Details of the recognition mechanism, including inferred global effects due to 4E10 binding, suggest that neutralization by 4E10 may involve mechanisms beyond simply binding, also requiring the ability of the antibody to induce conformational changes distant from its binding site. 4E10 is, therefore, unlikely to be re-elicited by conventional vaccination strategies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos , Anticuerpos Anti-VIH/inmunología , Anticuerpos Anti-VIH/metabolismo , Infecciones por VIH/inmunología , VIH-1/inmunología , Secuencia de Aminoácidos , Anticuerpos Monoclonales/química , Anticuerpos ampliamente neutralizantes , Cristalografía por Rayos X , Anticuerpos Anti-VIH/química , Infecciones por VIH/virología , Humanos , Datos de Secuencia Molecular , Pruebas de Neutralización , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
11.
Proc Natl Acad Sci U S A ; 110(20): E1867-76, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23630248

RESUMEN

We tested a strategy for engineering recombinant mammalian reoviruses (rMRVs) to express exogenous polypeptides. One important feature is that these rMRVs are designed to propagate autonomously and can therefore be tested in animals as potential vaccine vectors. The strategy has been applied so far to three of the 10 MRV genome segments: S3, M1, and L1. To engineer the modified segments, a 5' or 3' region of the essential, long ORF in each was duplicated, and then exogenous sequences were inserted between the repeats. The inner repeat and exogenous insert were positioned in frame with the native protein-encoding sequences but were separated from them by an in-frame "2A-like" sequence element that specifies a cotranslational "stop/continue" event releasing the exogenous polypeptide from the essential MRV protein. This design preserves a terminal region of the MRV genome segment with essential activities in RNA packaging, assortment, replication, transcription, and/or translation and alters the encoded MRV protein to a limited degree. Recovery of rMRVs with longer inserts was made more efficient by wobble-mutagenizing both the inner repeat and the exogenous insert, which possibly helped via respective reductions in homologous recombination and RNA structure. Immunogenicity of a 300-aa portion of the simian immunodeficiency virus Gag protein expressed in mice by an L1-modified rMRV was confirmed by detection of Gag-specific T-cell responses. The engineering strategy was further used for mapping the minimal 5'-terminal region essential to MRV genome segment S3.


Asunto(s)
Ingeniería Genética/métodos , Vectores Genéticos , Reoviridae/metabolismo , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Animales , Femenino , Productos del Gen gag/metabolismo , Genoma Viral , Ratones , Ratones Endogámicos C57BL , Conformación de Ácido Nucleico , Sistemas de Lectura Abierta , Péptidos/química , ARN Bicatenario/metabolismo , Virus de la Inmunodeficiencia de los Simios , Secuencias Repetidas en Tándem/genética
12.
PLoS Pathog ; 9(9): e1003639, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086134

RESUMEN

The broadly-neutralizing anti-HIV antibody 4E10 recognizes an epitope in the membrane-proximal external region of the HIV envelope protein gp41. Previous attempts to elicit 4E10 by vaccination with envelope-derived or reverse-engineered immunogens have failed. It was presumed that the ontogeny of 4E10-equivalent responses was blocked by inherent autoreactivity and exceptional polyreactivity. We generated 4E10 heavy-chain knock-in mice, which displayed significant B cell dysregulation, consistent with recognition of autoantigen/s by 4E10 and the presumption that tolerance mechanisms may hinder the elicitation of 4E10 or 4E10-equivalent responses. Previously proposed candidate 4E10 autoantigens include the mitochondrial lipid cardiolipin and a nuclear splicing factor, 3B3. However, using carefully-controlled assays, 4E10 bound only weakly to cardiolipin-containing liposomes, but also bound negatively-charged, non-cardiolipin-containing liposomes comparably poorly. 4E10/liposome binding was predominantly mediated by electrostatic interactions rather than presumed hydrophobic interactions. The crystal structure of 4E10 free of bound ligands showed a dramatic restructuring of the combining site, occluding the HIV epitope binding site and revealing profound flexibility, but creating an electropositive pocket consistent with non-specific binding of phospholipid headgroups. These results strongly suggested that antigens other than cardiolipin mediate 4E10 autoreactivity. Using a synthetic peptide library spanning the human proteome, we determined that 4E10 displays limited and focused, but unexceptional, polyspecificity. We also identified a novel autoepitope shared by three ER-resident inositol trisphosphate receptors, validated through binding studies and immunohistochemistry. Tissue staining with 4E10 demonstrated reactivity consistent with the type 1 inositol trisphosphate receptor as the most likely candidate autoantigen, but is inconsistent with splicing factor 3B3. These results demonstrate that 4E10 recognition of liposomes competes with MPER recognition and that HIV antigen and autoepitope recognition may be distinct enough to permit eliciting 4E10-like antibodies, evading autoimmunity through directed engineering. However, 4E10 combining site flexibility, exceptional for a highly-matured antibody, may preclude eliciting 4E10 by conventional immunization strategies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Regiones Determinantes de Complementariedad/inmunología , Epítopos/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Receptores de Inositol 1,4,5-Trifosfato/inmunología , Animales , Anticuerpos Monoclonales/genética , Autoanticuerpos/genética , Autoantígenos/genética , Anticuerpos ampliamente neutralizantes , Cardiolipinas/genética , Cardiolipinas/inmunología , Regiones Determinantes de Complementariedad/genética , Epítopos/genética , Anticuerpos Anti-VIH/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Ratones , Ratones Transgénicos , Proteoma/genética , Proteoma/inmunología
13.
Blood ; 122(3): 348-56, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23673862

RESUMEN

Many of the most promising tumor antigens for T-cell-based cancer immunotherapies are unmodified self-antigens. Unfortunately, the avidity of T cells specific for these antigens is limited by central tolerance during T-cell development in the thymus, resulting in decreased anti-tumor efficacy of these T cells. One approach to overcoming this obstacle is to mutate T-cell receptor (TCR) genes from naturally occurring T cells to enhance the affinity for the target antigen. These enhanced-affinity TCRs can then be developed for use in TCR gene therapy. Although TCRs with significantly enhanced affinity have been generated using this approach, it is not clear whether these TCRs, which bypass the affinity limits imposed by negative selection, remain unresponsive to the low levels of self-antigen generally expressed by some normal tissues. Here we show that 2 variants of a high-affinity WT1-specific TCR with enhanced affinity for WT1 are safe and do not mediate autoimmune tissue infiltration or damage when transduced into peripheral CD8 T cells and transferred in vivo. However, if expressed in developing T cells and subjected to thymic selection, the same enhanced-affinity TCRs signal tolerance mechanisms in the thymus, resulting in T cells with attenuated antigen sensitivity in the periphery.


Asunto(s)
Antígenos de Neoplasias/inmunología , Autoantígenos/inmunología , Terapia Genética , Receptores de Antígenos de Linfocitos T/inmunología , Timo/inmunología , Animales , Humanos , Listeria monocytogenes/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Mutantes/inmunología , Linfocitos T/inmunología , Transducción Genética
14.
J Allergy Clin Immunol ; 134(1): 195-203, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24698324

RESUMEN

BACKGROUND: Perforin-1 (Prf1) is the predominant cytolytic protein secreted by natural killer (NK) cells. For a rapid immune response, resting NK cells contain high Prf1 mRNA concentrations while exhibiting minimal cytotoxicity caused by a blockage of Prf1 protein synthesis, implying that an unknown posttranscriptional regulatory mechanism exists. OBJECTIVE: We sought to determine whether microRNA-150 (miR-150) posttranscriptionally regulates Prf1 translation in both mouse and human NK cells at rest and at various time points after activation. METHODS: Mouse NK cells with a targeted deletion of miR-150 (miR-150(-/-) NK cells), primary human NK cells, and NK92 MI cells were used to investigate the role of miR-150 in NK cells. NK cell cytotoxicity assays and Western blotting proved that activated miR-150(-/-) NK cells expressed upregulated Prf1, augmenting NK cell cytotoxicity. When immunodeficient mice were injected with miR-150(-/-) NK cells, there was a significant reduction in tumor growth and metastasis of B16F10 melanoma. RESULTS: We report that miR-150 binds to 3' untranslated regions of mouse and human Prf1, posttranscriptionally downregulating its expression. Mouse wild-type NK cells displayed downregulated miR-150 expression in response to IL-15, which led to corresponding repression and induction of Prf1 during rest and after IL-15 activation, respectively. CONCLUSION: Our results indicate that miR-150 is a common posttranscriptional regulator for Prf1 in mouse and human NK cells that represses NK cell lytic activity. Thus the therapeutic control of miR-150 in NK cells could enhance NK cell-based immunotherapy against cancer, providing a better clinical outcome.


Asunto(s)
Citotoxicidad Inmunológica/genética , Células Asesinas Naturales/inmunología , MicroARNs/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Regiones no Traducidas 3' , Animales , Sitios de Unión , Células Cultivadas , Regulación de la Expresión Génica/inmunología , Humanos , Interleucina-15/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/trasplante , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , MicroARNs/inmunología , Perforina , Proteínas Citotóxicas Formadoras de Poros/inmunología , Biosíntesis de Proteínas , Transducción de Señal , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia
15.
Gut ; 63(11): 1769-81, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24555999

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDA) is characterised by a robust desmoplasia, including the notable accumulation of immunosuppressive cells that shield neoplastic cells from immune detection. Immune evasion may be further enhanced if the malignant cells fail to express high levels of antigens that are sufficiently immunogenic to engender an effector T cell response. OBJECTIVE: To investigate the predominant subsets of immunosuppressive cancer-conditioned myeloid cells that chronicle and shape the progression of pancreas cancer. We show that selective depletion of one subset of myeloid-derived suppressor cells (MDSC) in an autochthonous, genetically engineered mouse model (GEMM) of PDA unmasks the ability of the adaptive immune response to engage and target tumour epithelial cells. METHODS: A combination of in vivo and in vitro studies were performed employing a GEMM that faithfully recapitulates the cardinal features of human PDA. The predominant cancer-conditioned myeloid cell subpopulation was specifically targeted in vivo and the biological outcomes determined. RESULTS: PDA orchestrates the induction of distinct subsets of cancer-associated myeloid cells through the production of factors known to influence myelopoiesis. These immature myeloid cells inhibit the proliferation and induce apoptosis of activated T cells. Targeted depletion of granulocytic MDSC (Gr-MDSC) in autochthonous PDA increases the intratumoral accumulation of activated CD8 T cells and apoptosis of tumour epithelial cells and also remodels the tumour stroma. CONCLUSIONS: Neoplastic ductal cells of the pancreas induce distinct myeloid cell subsets that promote tumour cell survival and accumulation. Targeted depletion of a single myeloid subset, the Gr-MDSC, can unmask an endogenous T cell response, disclosing an unexpected latent immunity and invoking targeting of Gr-MDSC as a potential strategy to exploit for treating this highly lethal disease.


Asunto(s)
Inmunidad Adaptativa , Carcinoma Ductal Pancreático/inmunología , Células Mieloides/inmunología , Neoplasias Pancreáticas/inmunología , Animales , Carcinoma Ductal Pancreático/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ingeniería Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Técnicas In Vitro , Ratones , Células Mieloides/patología , Mielopoyesis/inmunología , Neoplasias Pancreáticas/patología , Bazo/inmunología
16.
Carcinogenesis ; 35(7): 1451-60, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24908682

RESUMEN

Pancreatic ductal adenocarcinoma co-opts multiple cellular and extracellular mechanisms to create a complex cancer organ with an unusual proclivity for metastasis and resistance to therapy. Cell-autonomous events are essential for the initiation and maintenance of pancreatic ductal adenocarcinoma, but recent studies have implicated critical non-cell autonomous processes within the robust desmoplastic stroma that promote disease pathogenesis and resistance. Thus, non-malignant cells and associated factors are culprits in tumor growth, immunosuppression and invasion. However, even this increasing awareness of non-cell autonomous contributions to disease progression is tempered by the conflicting roles stromal elements can play. A greater understanding of stromal complexity and complicity has been aided in part by studies in highly faithful genetically engineered mouse models of pancreatic ductal adenocarcinoma. Insights gleaned from such studies are spurring the development of therapies designed to reengineer the pancreas cancer stroma and render it permissive to agents targeting cell-autonomous events or to reinstate immunosurveillance. Integrating conventional and immunological treatments in the context of stromal targeting may provide the key to a durable clinical impact on this formidable disease.


Asunto(s)
Matriz Extracelular/patología , Neoplasias Pancreáticas/patología , Células del Estroma/patología , Ingeniería de Tejidos , Animales , Humanos , Ratones
17.
Blood ; 119(23): 5492-501, 2012 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-22529286

RESUMEN

Targeted T-cell therapy is a potentially less toxic strategy than allogeneic stem cell transplantation for providing a cytotoxic antileukemic response to eliminate leukemic stem cells (LSCs) in acute myeloid leukemia (AML). However, this strategy requires identification of leukemia-associated antigens that are immunogenic and exhibit selective high expression in AML LSCs. Using microarray expression analysis of LSCs, hematopoietic cell subpopulations, and peripheral tissues to screen for candidate antigens, cyclin-A1 was identified as a candidate gene. Cyclin-A1 promotes cell proliferation and survival, has been shown to be leukemogenic in mice, is detected in LSCs of more than 50% of AML patients, and is minimally expressed in normal tissues with exception of testis. Using dendritic cells pulsed with a cyclin-A1 peptide library, we generated T cells against several cyclin-A1 oligopeptides. Two HLA A*0201-restricted epitopes were further characterized, and specific CD8 T-cell clones recognized both peptide-pulsed target cells and the HLA A*0201-positive AML line THP-1, which expresses cyclin-A1. Furthermore, cyclin-A1-specific CD8 T cells lysed primary AML cells. Thus, cyclin-A1 is the first prototypic leukemia-testis-antigen to be expressed in AML LSCs. The pro-oncogenic activity, high expression levels, and multitude of immunogenic epitopes make it a viable target for pursuing T cell-based therapy approaches.


Asunto(s)
Ciclina A1/genética , Ciclina A1/inmunología , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Células Madre Neoplásicas/inmunología , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Línea Celular Tumoral , Ciclina A1/análisis , Mapeo Epitopo , Antígeno HLA-A2/inmunología , Humanos , Masculino , Células Madre Neoplásicas/metabolismo , Testículo/inmunología
18.
Blood ; 119(17): 3940-50, 2012 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-22308288

RESUMEN

Cellular immune responses have the potential to elicit dramatic and sustained clinical remissions in lymphoma patients. Recent clinical trial data demonstrate that modification of T cells with chimeric antigen receptors (CARs) is a promising strategy. T cells containing CARs with costimulatory domains exhibit improved activity against tumors. We conducted a pilot clinical trial testing a "third-generation" CD20-specific CAR with CD28 and 4-1BB costimulatory domains in patients with relapsed indolent B-cell and mantle cell lymphomas. Four patients were enrolled, and 3 received T-cell infusions after cyclophosphamide lymphodepletion. Treatment was well tolerated, although one patient developed transient infusional symptoms. Two patients without evaluable disease remained progression-free for 12 and 24 months. The third patient had an objective partial remission and relapsed at 12 months after infusions. Modified T cells were detected by quantitative PCR at tumor sites and up to 1 year in peripheral blood, albeit at low levels. No evidence of host immune responses against infused cells was detected. In conclusion, adoptive immunotherapy with CD20-specific T cells was well tolerated and was associated with antitumor activity. We will pursue alternative gene transfer technologies and culture conditions in future studies to improve CAR expression and cell production efficiency.


Asunto(s)
Antígenos CD20/metabolismo , Antígenos CD28/genética , Inmunoterapia Adoptiva , Linfoma/terapia , Receptores de Antígenos/genética , Linfocitos T/trasplante , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Anciano , Anciano de 80 o más Años , Antígenos CD20/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígenos CD28/inmunología , Humanos , Linfoma/inmunología , Masculino , Persona de Mediana Edad , Proyectos Piloto , Pronóstico , Receptores de Antígenos/inmunología , Linfocitos T/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
19.
J Immunol ; 189(6): 3221-30, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22865917

RESUMEN

To understand better how selection processes balance the benefits of Ig repertoire diversity with the risks of autoreactivity and nonfunctionality of highly variable IgH CDR3s, we collected millions of rearranged germline IgH CDR3 sequences by deep sequencing of DNA from mature human naive B cells purified from four individuals and analyzed the data with computational methods. Long HCDR3 regions, often components of HIV-neutralizing Abs, appear to derive not only from incorporation of long D genes and insertion of large N regions but also by usage of multiple D gene segments in tandem. However, comparison of productive and out-of-frame IgH rearrangements revealed a selection bias against long HCDR3 loops, suggesting these may be disproportionately either poorly functional or autoreactive. Our data suggest that developmental selection removes HCDR3 loops containing patches of hydrophobicity, which are commonly found in some auto-antibodies, and at least 69% of the initial productive IgH rearrangements are removed from the repertoire during B cell development. Additionally, we have demonstrated the potential utility of this new technology for vaccine development with the identification in all four individuals of related candidate germline IgH precursors of the HIV-neutralizing Ab 4E10.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Reordenamiento Génico de Linfocito B/inmunología , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Análisis de Secuencia de ADN , Anticuerpos Neutralizantes/genética , Subgrupos de Linfocitos B/citología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Regiones Determinantes de Complementariedad/biosíntesis , Regiones Determinantes de Complementariedad/genética , Biología Computacional , Secuencia Conservada/genética , Secuencia Conservada/inmunología , VIH-1/genética , VIH-1/inmunología , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Región de Unión de la Inmunoglobulina/biosíntesis , Región de Unión de la Inmunoglobulina/genética , Región Variable de Inmunoglobulina/biosíntesis , Región Variable de Inmunoglobulina/genética , Precursores de Proteínas/biosíntesis , Precursores de Proteínas/genética , Análisis de Secuencia de ADN/métodos , Hipermutación Somática de Inmunoglobulina
20.
J Immunol ; 189(8): 3936-46, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22984076

RESUMEN

Adoptive T cell therapy (ACT) for the treatment of established cancers is actively being pursued in clinical trials. However, poor in vivo persistence and maintenance of antitumor activity of transferred T cells remain major problems. TGF-ß is a potent immunosuppressive cytokine that is often expressed at high levels within the tumor microenvironment, potentially limiting T cell-mediated antitumor activity. In this study, we used a model of autochthonous murine prostate cancer to evaluate the effect of cell-intrinsic abrogation of TGF-ß signaling in self/tumor-specific CD8 T cells used in ACT to target the tumor in situ. We found that persistence and antitumor activity of adoptively transferred effector T cells deficient in TGF-ß signaling were significantly improved in the cancerous prostate. However, over time, despite persistence in peripheral lymphoid organs, the numbers of transferred cells in the prostate decreased and the residual prostate-infiltrating T cells were no longer functional. These findings reveal that TGF-ß negatively regulates the accumulation and effector function of transferred self/tumor-specific CD8 T cells and highlight that, when targeting a tumor Ag that is also expressed as a self-protein, additional substantive obstacles are operative within the tumor microenvironment, potentially hampering the success of ACT for solid tumors.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Epítopos de Linfocito T/inmunología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/fisiología , Traslado Adoptivo/métodos , Animales , Linfocitos T CD8-positivos/trasplante , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neoplasias de la Próstata/terapia , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA