Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38559220

RESUMEN

Infiltration of immunosuppressive cells into the breast tumor microenvironment (TME) is associated with suppressed effector T cell (Teff) responses, accelerated tumor growth, and poor clinical outcomes. Previous studies from our group and others identified infiltration of immunosuppressive myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) as critical contributors to immune dysfunction in the orthotopic triple-negative breast cancer (TNBC) tumor model limiting the efficacy of adoptive cellular therapy. However, approaches to target these cells specifically in the TME are currently lacking. To overcome this barrier, polymeric micelles nanoparticles (PMNPs) were used for co-delivery of small molecule drugs activating Toll-like receptors 7 and 8 (TLR7/8) and inhibiting PI3K delta. The immunomodulation of the TME by TLR7/8 agonist and PI3K inhibitor altered macrophage polarization, reduced MDSC accumulation and selectively decreased tissue-resident Tregs in the TME, while enhancing the T and B cell adaptive immune response. PMNPs significantly enhanced the anti-tumor activity of local radiation therapy (RT) in mice bearing orthotopic TNBC tumors compared to RT alone. Taken together, these data demonstrate that RT combined with a nanoformulated immunostimulant restructured the TME and has promising potential for future translation combined with RT for patients with TNBC.

2.
J Immunol ; 211(2): 219-228, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37204246

RESUMEN

Previous work from our group and others has shown that patients with breast cancer can generate a T cell response against specific human epidermal growth factor 2 (HER2) epitopes. In addition, preclinical work has shown that this T cell response can be augmented by Ag-directed mAb therapy. This study evaluated the activity and safety of a combination of dendritic cell (DC) vaccination given with mAb and cytotoxic therapy. We performed a phase I/II study using autologous DCs pulsed with two different HER2 peptides given with trastuzumab and vinorelbine to a study cohort of patients with HER2-overexpressing and a second with HER2 nonoverexpressing metastatic breast cancer. Seventeen patients with HER2-overexpressing and seven with nonoverexpressing disease were treated. Treatment was well tolerated, with one patient removed from therapy because of toxicity and no deaths. Forty-six percent of patients had stable disease after therapy, with 4% achieving a partial response and no complete responses. Immune responses were generated in the majority of patients but did not correlate with clinical response. However, in one patient, who has survived >14 y since treatment in the trial, a robust immune response was demonstrated, with 25% of her T cells specific to one of the peptides in the vaccine at the peak of her response. These data suggest that autologous DC vaccination when given with anti-HER2-directed mAb therapy and vinorelbine is safe and can induce immune responses, including significant T cell clonal expansion, in a subset of patients.


Asunto(s)
Neoplasias de la Mama , Neoplasias Mamarias Animales , Humanos , Femenino , Animales , Epítopos/metabolismo , Vinorelbina/metabolismo , Vinorelbina/uso terapéutico , Receptor ErbB-2 , Neoplasias de la Mama/metabolismo , Inmunoterapia , Péptidos/metabolismo , Células Dendríticas , Trastuzumab/uso terapéutico , Trastuzumab/metabolismo
3.
Am J Transplant ; 23(2): 180-189, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36695691

RESUMEN

Belatacept-based immunosuppression in kidney transplantation confers fewer off-target toxicities than calcineurin inhibitors but comes at a cost of increased incidence and severity of acute rejection, potentially due to its deleterious effect on both the number and function of Foxp3+ regulatory T cells (Tregs). TIGIT is a CD28 family coinhibitory receptor expressed on several subsets of immune cells including Tregs. We hypothesized that coinhibition through TIGIT signaling could function to ameliorate costimulation blockade-resistant rejection. The results demonstrate that treatment with an agonistic anti-TIGIT antibody, when combined with costimulation blockade by CTLA-4Ig, can prolong allograft survival in a murine skin graft model compared with CTLA-4Ig alone. Further, this prolongation of graft survival is accompanied by an increase in the frequency and number of graft-infiltrating Tregs and a concomitant reduction in the number of CD8+ T cells in the graft. Through the use of Treg-specific TIGIT conditional knockout animals, we demonstrated that the TIGIT-mediated reduction in the graft-infiltrating CD8+ T cell response is dependent on signaling of TIGIT on Foxp3+ Tregs. Our results highlight both the key functional role of TIGIT on Foxp3+ Tregs under conditions in which CTLA-4 is blocked and the therapeutic potential of TIGIT agonism to optimize costimulation blockade-based immunosuppression.


Asunto(s)
Trasplante de Riñón , Linfocitos T Reguladores , Animales , Ratones , Abatacept/farmacología , Linfocitos T CD8-positivos , Factores de Transcripción Forkhead , Rechazo de Injerto/tratamiento farmacológico , Rechazo de Injerto/etiología , Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Trasplante de Riñón/efectos adversos , Trasplante Homólogo
4.
Blood Adv ; 5(22): 4578-4589, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34619767

RESUMEN

Acute graft-versus-host disease (aGVHD), mediated by the recognition of host major histocompatibility complex/peptide polymorphisms by donor T cells, remains a significant complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). aGVHD most commonly involves the gastrointestinal tract, liver, and skin; symptomatic aGVHD is treated with corticosteroids. Steroid-nonresponsive aGVHD is a significant problem for patients undergoing allo-HSCT, with <15% of these patients alive 1 year after diagnosis. Previously, we found that the infusion of donor innate lymphoid type 2 (ILC2) cells could prevent and treat aGVHD of the lower gastrointestinal tract with no effect on the graft-versus-leukemia response. This approach for clinical translation is cumbersome, as it would require the generation of donor-derived ILC2 cells for each recipient. Thus, the ability to use third-party ILC2 cells would provide an "off-the-shelf" reagent that could be used to treat and/or prevent aGVHD. Here, we show that third-party ILC2 cells enhance the survival of allo-HSCT recipients. Treatment required at least 4 weekly infusions of ILC2 cells. Mechanistically, we show that ILC2 cell function was completely lost if the cells could not express both interleukin-13 (IL-13) and amphiregulin. Finally, we show that the activity of IL-13 has a greater dependence on the expression of the IL-13R on host rather than donor bone marrow cells. The ability to generate third-party ILC2 cells offers a new avenue for the prevention of aGVHD.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Tracto Gastrointestinal , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Inmunidad Innata , Linfocitos
5.
J Exp Med ; 218(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33382402

RESUMEN

CAR T therapy targeting solid tumors is restrained by limited infiltration and persistence of those cells in the tumor microenvironment (TME). Here, we developed approaches to enhance the activity of CAR T cells using an orthotopic model of locally advanced breast cancer. CAR T cells generated from Th/Tc17 cells given with the STING agonists DMXAA or cGAMP greatly enhanced tumor control, which was associated with enhanced CAR T cell persistence in the TME. Using single-cell RNA sequencing, we demonstrate that DMXAA promoted CAR T cell trafficking and persistence, supported by the generation of a chemokine milieu that promoted CAR T cell recruitment and modulation of the immunosuppressive TME through alterations in the balance of immune-stimulatory and suppressive myeloid cells. However, sustained tumor regression was accomplished only with the addition of anti-PD-1 and anti-GR-1 mAb to Th/Tc17 CAR T cell therapy given with STING agonists. This study provides new approaches to enhance adoptive T cell therapy in solid tumors.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas de la Membrana/agonistas , Receptores Quiméricos de Antígenos/metabolismo , Células 3T3 , Animales , Línea Celular , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Inmunoterapia Adoptiva/métodos , Ratones , Linfocitos T/metabolismo , Microambiente Tumoral/fisiología
6.
JCI Insight ; 4(9)2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31045575

RESUMEN

Recent seminal studies have revealed that laboratory mice differ from adult humans with regard to the frequency, number, and distribution of memory T cells. Because our data show that memory T cells are more susceptible to sepsis-induced death than naive T cells, in this study we developed a model in which mice possess a memory T cell compartment more similar to that of adult humans, to better study immune responses during sepsis in the more physiologically relevant context of high frequencies of memory T cells. Using this model, we found that CD44hi memory T cells significantly upregulated the coinhibitory molecule 2B4 during sepsis, and 2B4+ memory T cells coexpressed markers of both activation and exhaustion. Genetic deficiency in 2B4 resulted in decreased mortality during sepsis. Mechanistically, this decreased mortality was associated with reduced caspase-3/7+ apoptotic T cells in 2B4-/- relative to WT, septic hosts. These results were corroborated by analysis of PBMCs isolated from human patients with sepsis, which showed increased frequencies of caspase-3/7+ apoptotic cells among 2B4+ relative to 2B4- T cells. Thus, 2B4 plays a critical role in sepsis-induced apoptosis in both murine memory T cells and those isolated from human patients with sepsis.


Asunto(s)
Memoria Inmunológica/inmunología , Sepsis/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Linfocitos T/inmunología , Adulto , Animales , Apoptosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Humanos , Receptores de Hialuranos/inmunología , Estimación de Kaplan-Meier , Listeria monocytogenes , Listeriosis , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Sepsis/microbiología , Sepsis/mortalidad , Transducción de Señal , Regulación hacia Arriba
7.
J Immunol ; 201(5): 1536-1548, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30012849

RESUMEN

We recently showed that 2B4 expression on memory T cells in human renal transplant recipients was associated with reduced rates of rejection. To investigate whether 2B4 functionally underlies graft acceptance during transplantation, we established an experimental model in which 2B4 was retrogenically expressed on donor-reactive murine CD8+ T cells (2B4rg), which were then transferred into naive recipients prior to skin transplantation. We found that constitutive 2B4 expression resulted in significantly reduced accumulation of donor-reactive CD8+ T cells following transplantation and significantly prolonged graft survival following transplantation. This marked reduction in alloreactivity was due to reduced proliferation of CD8+ Thy1.1+ 2B4rg cells as compared with control cells, underpinned by extracellular flux analyses demonstrating that 2B4-deficient (2B4KO) CD8+ cells activated in vitro exhibited increased glycolytic capacity and upregulation of gene expression profiles consistent with enhanced glycolytic machinery as compared with wild type controls. Furthermore, 2B4KO CD8+ T cells primed in vivo exhibited significantly enhanced ex vivo uptake of a fluorescent glucose analogue. Finally, the proliferative advantage associated with 2B4 deficiency was only observed in the setting of glucose sufficiency; in glucose-poor conditions, 2B4KO CD8+ T cells lost their proliferative advantage. Together, these data indicate that 2B4 signals function to alter T cell glucose metabolism, thereby limiting the proliferation and accumulation of CD8+ T cells. Targeting 2B4 may therefore represent a novel therapeutic strategy to attenuate unwanted CD8+ T cell responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , División Celular/inmunología , Glucólisis/inmunología , Supervivencia de Injerto/inmunología , Activación de Linfocitos , Transducción de Señal/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Trasplante de Piel , Animales , División Celular/genética , Glucólisis/genética , Supervivencia de Injerto/genética , Ratones , Ratones Noqueados , Transducción de Señal/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética
8.
Transpl Immunol ; 50: 34-42, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29885905

RESUMEN

Models of infection have shaped our understanding of programmed memory T cell differentiation, yet whether these models apply to memory programming in the context of transplantation has yet to be defined. Previous work has identified differences in the response of antigen-specific CD8+ T cells to cognate antigen based on the environment in which the antigen is presented. Thus, we hypothesized that programming of antigen specific CD8+ T cells responding to graft and pathogen may be dissimilar. Here we find that antigen-specific CD8+ T cells primed by a skin graft contract faster than those primed by gammaherpesvirus (gHV), yet are able to expand more rapidly upon rechallenge. Moreover, graft-primed antigen-specific CD8+ T cells exhibited higher frequencies of cells secreting IL-2 and demonstrate lower expression of KLRG-1, which are qualities suggestive of increased recall potential. Additionally, the expression of CD127 at a memory time point suggests graft-elicited CD8+ antigen specific T cells are maintained in a less terminally-differentiated state compared to gHV-elicited CD8+ antigen specific T cells, despite fewer cells being present at that time point. Taken together, our findings suggest that the surface marker expression and functional profiles of T cells depends on the priming conditions and may be used to predict immunologic risk following transplantation after traditional allosensitization or heterologous immune priming.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Gammaherpesvirinae/inmunología , Rechazo de Injerto/inmunología , Infecciones por Herpesviridae/inmunología , Trasplante de Piel , Animales , Antígenos Virales/inmunología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Memoria Inmunológica , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Isoantígenos/inmunología , Lectinas Tipo C , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Inmunológicos/metabolismo
10.
J Nutrigenet Nutrigenomics ; 5(2): 81-93, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22722880

RESUMEN

BACKGROUND/AIMS: Recent studies have highlighted the importance of gene by diet interactions in contributing to risk factors of metabolic syndrome. We used a consomic rat panel, in which a chromosome of the Brown Norway (BN) strain is introgressed onto the background of the Dahl salt-sentitive (SS) strain, to test the hypothesis that these animals will be useful for dissecting gene by diet interactions involved in metabolic syndrome. METHODS: We placed the parental SS and BN strains on a low-fat/high-carbohydrate (LF) or high-fat/low-carbohydrate (HF) diet for 22 weeks and measured several indices of metabolic syndrome. We then investigated the effect of diet in eight consomic rat strains. RESULTS: We show that the HF diet resulted in significantly increased levels of fasting plasma cholesterol and triglycerides in the SS strain, with no effect in the BN. Both strains responded to the HF diet with slight increases in body weight. SSBN8 was the only consomic strain that resembled that of the BN, with low levels of fasting cholesterol and triglycerides even on the HF diet. CONCLUSIONS: These results indicate that BN chromosome 8 harbors a gene or genes that confer protection against dyslipidemia caused by the HF diet.


Asunto(s)
Mapeo Cromosómico , Carbohidratos de la Dieta/administración & dosificación , Grasas de la Dieta/administración & dosificación , Dislipidemias/prevención & control , Animales , Dislipidemias/genética , Masculino , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...