Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur J Immunol ; 50(9): 1386-1399, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32383488

RESUMEN

Adoptive T cell transfer therapy induces objective responses in patients with advanced malignancies. Despite these results, some individuals do not respond due to the generation of terminally differentiated T cells during the expansion protocol. As the gamma and delta catalytic subunits in the PI3K pathway are abundant in leukocytes and involved in cell activation, we posited that blocking both subunits ex vivo with the inhibitor IPI-145 would prevent their differentiation, thereby increasing antitumor activity in vivo. However, IPI-145 treatment generated a product with reduced antitumor activity. Instead, T cells inhibited of PI3Kγ (IPI-549) or PI3Kδ (CAL-101 or TGR-1202) alone were more potent in vivo. While T cells coinhibited of PI3Kγ and PI3Kδ were less differentiated, they were functionally impaired, indicated by reduced production of effector cytokines after antigenic re-encounter and decreased persistence in vivo. Human CAR T cells expanded with either a PI3Kγ or PI3Kδ inhibitor possessed a central memory phenotype compared to vehicle cohorts. We also found that PI3Kδ-inhibited CARs lysed human tumors in vitro more effectively than PI3Kγ-expanded or traditionally expanded CAR T cells. Our data imply that sole blockade of PI3Kγ or PI3Kδ generates T cells with remarkable antitumor properties, a discovery that has substantial clinical implications.


Asunto(s)
Linfocitos T CD8-positivos/trasplante , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Inmunoterapia Adoptiva/métodos , Animales , Fosfatidilinositol 3-Quinasa Clase I/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase Ib/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Isoquinolinas/farmacología , Ratones , Pirazoles/farmacología , Pirimidinas/farmacología , Receptores Quiméricos de Antígenos
2.
Cancer Immunol Immunother ; 70(5): 1165-1175, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33104836

RESUMEN

The solid tumor microenvironment is replete with factors that present a stress to infiltrating immune cells. Endoplasmic reticulum (ER) stress sensor PKR-like ER kinase (PERK) is primed to sense and respond to the burden of misfolded proteins in the ER lumen induced by cell stressors. PERK has documented roles as a master regulator of acute and chronic responses to cell stress as well as in the regulation of cell metabolism. Here, we provide an overview of the roles of PERK based on what is known and remains to be tested in immune cells in tumors and impacts on tumor control. PERK is one of several ER kinases able to preferentially induce activating transcription factor 4 (ATF4) as a response to cell stress. ATF4 orchestrates the oxidative stress response and governs amino acid metabolism. We discuss the tested role of ATF4 in tumor immunity and provide insight on the dueling protective and deleterious roles that ATF4 may play in the stress of solid tumors.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Estrés del Retículo Endoplásmico/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Linfocitos T/inmunología , Factor de Transcripción Activador 4/genética , Animales , Humanos , Inmunidad , Microambiente Tumoral , eIF-2 Quinasa/metabolismo
3.
J Immunol ; 192(12): 5679-86, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24808359

RESUMEN

CD24 binds to and suppresses inflammation triggered by danger-associated molecular patterns such as heat shock proteins (HSPs) and high-mobility group box 1. Paradoxically, CD24 has been shown to enhance autoimmune disease. In this study, we attempt to reconcile this paradox by deletion of CD24 (24KO) in a lupus-like disease model driven by forced expression of HSP gp96 at the cell surface (transgenic mice [tm]). As expected, tm24KO mice showed increased CD11c(+) dendritic cell activation coupled to a significant increase in dendritic cell-specific IL-12 production compared with tm mice. However, tm24KO mice showed less CD4 T cell activation and peripheral inflammatory cytokine production in comparison with tm mice. We characterized an enhanced immune suppressive milieu in tm24KO mice distinguished by increased TGF-ß and greater regulatory T cell-suppressive capacity. We found greater absolute numbers of myeloid-derived suppressor cells (MDSCs) in tm24KO mice and showed that the Ly6C(+) MDSC subset had greater suppressive capacity from tm24KO mice. Deletion of CD24 in tm mice led to diminished lupus-like pathology as evidenced by anti-nuclear Ab deposition and glomerulonephritis. Finally, we show that expanded MDSC populations were mediated by increased free high-mobility group box 1 in tm24KO mice. Thus, the deletion of CD24 in an HSP-driven model of autoimmunity led to the unexpected development of regulatory T cell and MDSC populations that augmented immune tolerance. Further study of these populations as possible negative regulators of inflammation in the context of autoimmunity is warranted.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Antígeno CD24/inmunología , Células Dendríticas/inmunología , Eliminación de Gen , Glicoproteínas de Membrana/inmunología , Células Mieloides/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Antígeno CD11c/genética , Antígeno CD11c/inmunología , Antígeno CD24/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Células Dendríticas/patología , Tolerancia Inmunológica/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Células Mieloides/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
4.
J Immunol ; 190(7): 3639-47, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23455498

RESUMEN

Infection and inflammation can disturb immune tolerance at the maternal-fetal interface, resulting in adverse pregnancy outcomes. However, the underlying mechanisms for detrimental immune responses remain ill defined. In this study, we provide evidence for immune programming of fetal loss in response to polyinosinic:polycytidylic acid (polyI:C), a viral mimic and an inducer of inflammatory milieu. IL-10 and uterine NK (uNK) cells expressing the activating receptor NKG2D play a critical role in poly(I:C)-induced fetal demise. In wild type (WT) mice, poly(I:C) treatment induced expansion of NKG2D(+) uNK cells and expression of Rae-1 (an NKG2D ligand) on uterine macrophages and led to fetal resorption. In IL-10(-/-) mice, NKG2D(-) T cells instead became the source of fetal resorption during the same gestation period. Interestingly, both uterine NK and T cells produced TNF-α as the key cytotoxic factor contributing to fetal loss. Treatment of WT mice with poly(I:C) resulted in excessive trophoblast migration into the decidua and increased TUNEL-positive signal. IL-10(-/-) mice supplemented with recombinant IL-10 induced fetal loss through NKG2D(+) uNK cells, similar to the response in WT mice. Blockade of NKG2D in poly(I:C)-treated WT mice led to normal pregnancy outcome. Thus, we demonstrate that pregnancy-disrupting inflammatory events mimicked by poly(I:C) are regulated by IL-10 and depend on the effector function of uterine NKG2D(+) NK cells in WT mice and NKG2D(-) T cells in IL-10 null mice.


Asunto(s)
Pérdida del Embrión/inducido químicamente , Pérdida del Embrión/genética , Interleucina-10/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/antagonistas & inhibidores , Poli I-C/efectos adversos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Pérdida del Embrión/prevención & control , Femenino , Reabsorción del Feto/inducido químicamente , Reabsorción del Feto/inmunología , Interleucina-10/deficiencia , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Embarazo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Regulación hacia Arriba , Útero/efectos de los fármacos , Útero/inmunología , Útero/metabolismo
5.
Cancer Res ; 84(1): 69-83, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-37801615

RESUMEN

Generating stem-like memory T cells (TSCM) is a potential strategy to improve adoptive immunotherapy. Elucidating optimal ways to modulate signaling pathways that enrich TSCM properties could identify approaches to achieve this goal. We discovered herein that blocking the PI3Kδ pathway pharmaceutically to varying degrees can generate T cells with increasingly heightened stemness properties, based on the progressive enrichment of the transcription factors Tcf1 and Lef1. T cells with enhanced stemness features exhibited metabolic plasticity, marked by improved mitochondrial function and glucose uptake after tumor recognition. Conversely, T cells with low or medium stemness were less metabolically dynamic, vulnerable to antigen-induced cell death, and expressed more inhibitory checkpoint receptors. Only T-cell receptor-specific or chimeric antigen receptor (CAR)-specific T cells with high stemness persisted in vivo and mounted protective immunity to tumors. Likewise, the strongest level of PI3Kδ blockade in vitro generated human tumor-infiltrating lymphocytes and CAR T cells with elevated stemness properties, in turn bolstering their capacity to regress human solid tumors. The stemness level of T cells in vitro was important, ultimately impacting their efficacy in mice bearing three distinct solid tumors. Lef1 and Tcf1 sustained antitumor protection by donor high CD8+ TSCM or CD4+ Th17SCM, as deletion of either one compromised the therapeutic efficacy. Collectively, these findings highlight the importance of strategic modulation of PI3Kδ signaling in T cells to induce stemness and lasting protective responses to solid tumors. SIGNIFICANCE: Elevating T-cell stemness by progressively blocking PI3Kδ signaling during ex vivo manufacturing of adoptive cell therapies alters metabolic and functional properties to enhance antitumor immunity dependent on Tcf1 and Lef1.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Ratones , Animales , Inmunoterapia Adoptiva , Linfocitos Infiltrantes de Tumor , Receptores de Antígenos de Linfocitos T , Linfocitos T CD8-positivos
6.
Cell Metab ; 36(5): 969-983.e10, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38490211

RESUMEN

The solid tumor microenvironment (TME) imprints a compromised metabolic state in tumor-infiltrating T cells (TILs), hallmarked by the inability to maintain effective energy synthesis for antitumor function and survival. T cells in the TME must catabolize lipids via mitochondrial fatty acid oxidation (FAO) to supply energy in nutrient stress, and it is established that T cells enriched in FAO are adept at cancer control. However, endogenous TILs and unmodified cellular therapy products fail to sustain bioenergetics in tumors. We reveal that the solid TME imposes perpetual acetyl-coenzyme A (CoA) carboxylase (ACC) activity, invoking lipid biogenesis and storage in TILs that opposes FAO. Using metabolic, lipidomic, and confocal imaging strategies, we find that restricting ACC rewires T cell metabolism, enabling energy maintenance in TME stress. Limiting ACC activity potentiates a gene and phenotypic program indicative of T cell longevity, engendering T cells with increased survival and polyfunctionality, which sustains cancer control.


Asunto(s)
Acetil-CoA Carboxilasa , Linfocitos T CD8-positivos , Metabolismo de los Lípidos , Microambiente Tumoral , Acetil-CoA Carboxilasa/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Ratones , Ratones Endogámicos C57BL , Humanos , Ácidos Grasos/metabolismo , Femenino , Línea Celular Tumoral , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Mitocondrias/metabolismo
7.
bioRxiv ; 2023 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-36945574

RESUMEN

Generating stem memory T cells (T SCM ) is a key goal for improving cancer immunotherapy. Yet, the optimal way to modulate signaling pathways that enrich T SCM properties remains elusive. Here, we discovered that the degree to which the PI3Kδ pathway is blocked pharmaceutically can generate T cells with differential levels of stemness properties. This observation was based on the progressive enrichment of transcriptional factors of stemness (Tcf-1 and Lef-1). Additional investigation revealed that T cells with high stemness features had enhanced metabolic plasticity, marked by heightened mitochondrial function and glucose uptake. Conversely, T cells with low or medium features of stemness expressed more inhibitory checkpoint receptors (Tim-3, CD39) and were vulnerable to antigen-induced cell death. Only TCR-antigen specific T cells with high stemness persisted following adoptive transfer in vivo and mounted protective immunity to melanoma tumors. Likewise, the strongest level of PI3Kδ blockade in vitro generated human tumor infiltrating lymphocytes (TILs) and CAR T cells with heightened stemness properties, in turn bolstering their capacity to regress human mesothelioma tumors. We find that the level of stemness T cells possess in vitro differentially impacts their potency upon transfer in three tumor models. Mechanistically, both Lef-1 and Tcf-1 sustain anti-tumor protection by high T SCM , as deletion of either one compromised cellular therapy. Collectively, these findings highlight the therapeutic potential of carefully modulating PI3Kδ signaling in T cells to confer high stemness and mediate protective responses to solid tumors.

8.
Front Cell Dev Biol ; 10: 867341, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35573704

RESUMEN

The endoplasmic reticulum (ER) is a large continuous membranous organelle that plays a central role as the hub of protein and lipid synthesis while the mitochondria is the principal location for energy production. T cells are an immune subset exhibiting robust dependence on ER and mitochondrial function based on the need for protein synthesis and secretion and metabolic dexterity associated with foreign antigen recognition and cytotoxic effector response. Intimate connections exist at mitochondrial-ER contact sites (MERCs) that serve as the structural and biochemical platforms for cellular metabolic homeostasis through regulation of fission and fusion as well as glucose, Ca2+, and lipid exchange. Work in the tumor immunotherapy field indicates that the complex interplay of nutrient deprivation and tumor antigen stimulation in the tumor microenvironment places stress on the ER and mitochondria, causing dysfunction in organellar structure and loss of metabolic homeostasis. Here, we assess prior literature that establishes how the structural interface of these two organelles is impacted by the stress of solid tumors along with recent advances in the manipulation of organelle homeostasis at MERCs in T cells. These findings provide strong evidence for increased tumor immunity using unique therapeutic avenues that recharge cellular metabolic homeostasis in T cells.

9.
Cancer Res ; 82(23): 4386-4399, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36126165

RESUMEN

Protein synthesis supports robust immune responses. Nutrient competition and global cell stressors in the tumor microenvironment (TME) may impact protein translation in T cells and antitumor immunity. Using human and mouse tumors, we demonstrated here that protein translation in T cells is repressed in solid tumors. Reduced glucose availability to T cells in the TME led to activation of the unfolded protein response (UPR) element eIF2α (eukaryotic translation initiation factor 2 alpha). Genetic mouse models revealed that translation attenuation mediated by activated p-eIF2α undermines the ability of T cells to suppress tumor growth. Reprograming T-cell metabolism was able to alleviate p-eIF2α accumulation and translational attenuation in the TME, allowing for sustained protein translation. Metabolic and pharmacological approaches showed that proteasome activity mitigates induction of p-eIF2α to support optimal antitumor T-cell function, protecting from translation attenuation and enabling prolonged cytokine synthesis in solid tumors. Together, these data identify a new therapeutic avenue to fuel the efficacy of tumor immunotherapy. SIGNIFICANCE: Proteasome function is a necessary cellular component for endowing T cells with tumor killing capacity by mitigating translation attenuation resulting from the unfolded protein response induced by stress in the tumor microenvironment.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Ratones , Animales , Linfocitos T/metabolismo , Complejo de la Endopetidasa Proteasomal , Neoplasias/terapia , Microambiente Tumoral , Inmunoterapia/métodos , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo
10.
J Immunother Cancer ; 10(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35017148

RESUMEN

BACKGROUND: Adoptive T cell transfer (ACT) therapy improves outcomes in patients with advanced malignancies, yet many individuals relapse due to the infusion of T cells with poor function or persistence. Toll-like receptor (TLR) agonists can invigorate antitumor T cell responses when administered directly to patients, but these responses often coincide with toxicities. We posited that TLR agonists could be repurposed ex vivo to condition T cells with remarkable potency in vivo, circumventing TLR-related toxicity. METHODS: In this study we investigated how tumor-specific murine CD8+ T cells and human tumor infiltrating lymphocytes (TILs) are impacted when expanded ex vivo with the TLR9 agonist CpG. RESULTS: Herein we reveal a new way to reverse the tolerant state of adoptively transferred CD8+ T cells against tumors using TLR-activated B cells. We repurposed the TLR9 agonist, CpG, commonly used in the clinic, to bolster T cell-B cell interactions during expansion for ACT. T cells expanded ex vivo from a CpG-treated culture demonstrated potent antitumor efficacy and prolonged persistence in vivo. This antitumor efficacy was accomplished without in vivo administration of TLR agonists or other adjuvants of high-dose interleukin (IL)-2 or vaccination, which are classically required for effective ACT therapy. CpG-conditioned CD8+ T cells acquired a unique proteomic signature hallmarked by an IL-2RαhighICOShighCD39low phenotype and an altered metabolic profile, all reliant on B cells transiently present in the culture. Likewise, human TILs benefitted from expansion with CpG ex vivo, as they also possessed the IL-2RαhighICOShighCD39low phenotype. CpG fostered the expansion of potent CD8+ T cells with the signature phenotype and antitumor ability via empowering a direct B-T cell interaction. Isolated B cells also imparted T cells with the CpG-associated phenotype and improved tumor immunity without the aid of additional antigen-presenting cells or other immune cells in the culture. CONCLUSIONS: Our results demonstrate a novel way to use TLR agonists to improve immunotherapy and reveal a vital role for B cells in the generation of potent CD8+ T cell-based therapies. Our findings have immediate implications in the clinical treatment of advanced solid tumors.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma/tratamiento farmacológico , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
11.
J Immunol ; 183(2): 1144-54, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19561095

RESUMEN

Pregnancy outcome is severely compromised by intrauterine infections and inflammation. Although the pregnant uterine microenvironment is replete with innate immune cells and TLR expression, the mechanisms that facilitate adverse effects of their activation are largely unknown. In this study, we mimic the activation of TLR9 with its pathogenic ligand hypomethylated CpG and demonstrate that IL-10 proficiency protects against CpG-induced pregnancy complications. We show that fetal resorption and preterm birth are rapidly induced in IL-10(-/-) mice by low doses of CpG (approximately 25 microg/mouse) when injected i.p. on gestational day 6 or gestational day 14, respectively. In contrast, wild-type mice failed to experience such effects at comparable doses, but pups born at term displayed craniofacial/limb defects in response to higher doses (approximately 400 microg/mouse). Pregnancy complications in IL-10(-/-) mice were associated with unexpected and robust TLR9-triggered activation and amplification of uterine neutrophil and macrophage subpopulations followed by their migration to the placental zone. Furthermore, a dramatic increase in serum levels of mouse KC and TNF-alpha production by uterine F4/80(+) cells, but not uterine NK or Gr-1(+)CD11b(+) cells, was observed. Depletion of F4/80(+) macrophages or neutralization of TNF-alpha rescued pregnancy to term. Our results have important implications for IL-10-mediated "uterine tolerance" against CpG-driven innate immune activation.


Asunto(s)
Interleucina-10/deficiencia , Complicaciones del Embarazo/etiología , Receptor Toll-Like 9/metabolismo , Animales , Huesos/anomalías , Femenino , Reabsorción del Feto , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/patología , Embarazo , Complicaciones del Embarazo/inducido químicamente , Resultado del Embarazo , Nacimiento Prematuro , Factor de Necrosis Tumoral alfa/biosíntesis , Útero/inmunología , Útero/metabolismo , Útero/patología
12.
Front Immunol ; 12: 645242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815400

RESUMEN

Emerging reports show that metabolic pathways can be targeted to enhance T cell-mediated immunity to tumors. Yet, tumors consume key metabolites in the host to survive, thus robbing T cells of these nutrients to function and thrive. T cells are often deprived of basic building blocks for energy in the tumor, including glucose and amino acids needed to proliferate or produce cytotoxic molecules against tumors. Immunosuppressive molecules in the host further compromise the lytic capacity of T cells. Moreover, checkpoint receptors inhibit T cell responses by impairing their bioenergetic potential within tumors. In this review, we discuss the fundamental metabolic pathways involved in T cell activation, differentiation and response against tumors. We then address ways to target metabolic pathways to improve the next generation of immunotherapies for cancer patients.


Asunto(s)
Metabolismo Energético/inmunología , Activación de Linfocitos , Neoplasias , Linfocitos T , Microambiente Tumoral/inmunología , Humanos , Inmunoterapia , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
13.
Cancer Prev Res (Phila) ; 14(9): 885-892, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34341013

RESUMEN

Immune responses vary in colorectal cancers, which strongly influence prognosis. However, little is known about the variance in immune response within preinvasive lesions. The study aims to investigate how the immune contexture differs by clinicopathologic features (location, histology, dysplasia) associated with progression and recurrence in early carcinogenesis. We performed a cross-sectional study using preinvasive lesions from the surgical pathology laboratory at the Medical University of South Carolina. We stained the tissues with immunofluorescence antibodies, then scanned and analyzed expression using automated image analysis software. We stained CD117 as a marker of mast cells, CD4/RORC to indicate Th17 cells, MICA/B as a marker of NK-cell ligands, and also used antibodies directed against cytokines IL6, IL17A, and IFNγ. We used negative binomial regression analysis to compare analyte density counts by location, histology, degree of dysplasia adjusted for age, sex, race, and batch. All immune markers studied (except IL17a) had significantly higher density counts in the proximal colon than distal colon and rectum. Increases in villous histology were associated with significant decreases in immune responses for IL6, IL17a, NK ligand, and mast cells. No differences were observed in lesions with low- and high-grade dysplasia, except in mast cells. The lesions of the proximal colon were rich in immune infiltrate, paralleling the responses observed in normal mucosa and invasive disease. The diminishing immune response with increasing villous histology suggests an immunologically suppressive tumor environment. Our findings highlight the heterogeneity of the immune responses in preinvasive lesions, which may have implications for prevention strategies. PREVENTION RELEVANCE: Our study is focused on immune infiltrate expression in preinvasive colorectal lesions; our results suggest important differences by clinicopathologic features that have implications for immune prevention research.


Asunto(s)
Adenoma/patología , Neoplasias Colorrectales/patología , Inmunidad/fisiología , Adenoma/diagnóstico , Adenoma/inmunología , Adenoma/metabolismo , Anciano , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Colon/inmunología , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Estudios Transversales , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Microambiente Tumoral/inmunología
14.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33653801

RESUMEN

Immune checkpoint inhibitors (ICIs) have improved overall survival for cancer patients, however, optimal duration of ICI therapy has yet to be defined. Given ICIs were first used to treat patients with metastatic melanoma, a condition that at the time was incurable, little attention was initially paid to how much therapy would be needed for a durable response. As the early immunotherapy trials have matured past 10 years, a significant per cent of patients have demonstrated durable responses; it is now time to determine whether patients have been overtreated, and if durable remissions can still be achieved with less therapy, limiting the physical and financial toxicity associated with years of treatment. Well-designed trials are needed to identify optimal duration of therapy, and to define biomarkers to predict who would benefit from shorter courses of immunotherapy. Here, we outline key questions related to health, financial and societal toxicities of over treating with ICI and present four unique clinical trials aimed at exposing criteria for early cessation of ICI. Taken together, there is a serious liability to overtreating patients with ICI and future work is warranted to determine when it is safe to stop ICI.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Neoplasias/tratamiento farmacológico , Ensayos Clínicos como Asunto , Esquema de Medicación , Medicina Basada en la Evidencia , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Neoplasias/inmunología , Neoplasias/mortalidad , Neoplasias/patología , Seguridad del Paciente , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento
15.
Nat Cancer ; 2(1): 66-82, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33738458

RESUMEN

Despite objective responses to PARP inhibition and improvements in progression-free survival compared to standard chemotherapy in patients with BRCA-associated triple-negative breast cancer (TNBC), benefits are transitory. Using high dimensional single-cell profiling of human TNBC, here we demonstrate that macrophages are the predominant infiltrating immune cell type in BRCA-associated TNBC. Through multi-omics profiling we show that PARP inhibitors enhance both anti- and pro-tumor features of macrophages through glucose and lipid metabolic reprogramming driven by the sterol regulatory element-binding protein 1 (SREBP-1) pathway. Combined PARP inhibitor therapy with CSF-1R blocking antibodies significantly enhanced innate and adaptive anti-tumor immunity and extends survival in BRCA-deficient tumors in vivo and is mediated by CD8+ T-cells. Collectively, our results uncover macrophage-mediated immune suppression as a liability of PARP inhibitor treatment and demonstrate combined PARP inhibition and macrophage targeting therapy induces a durable reprogramming of the tumor microenvironment, thus constituting a promising therapeutic strategy for TNBC.


Asunto(s)
Inhibidores de Poli(ADP-Ribosa) Polimerasas , Neoplasias de la Mama Triple Negativas , Proteína BRCA1/genética , Linfocitos T CD8-positivos , Línea Celular Tumoral , Humanos , Macrófagos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Microambiente Tumoral
16.
Artículo en Inglés | MEDLINE | ID: mdl-20827416

RESUMEN

The incidence of preterm birth in developed countries has risen in the past decades. Underlying causes for this enigmatic pregnancy complication are numerous, yet infectious agents that induce dysregulation of immunity at the maternal-fetal interface pose one of the most probable causes of preterm birth. This paper highlights two factors regarding maternal infections that trigger unscheduled inflammatory sequences that are deleterious to the maternal-fetal balance necessary to maintain pregnancy. Firstly, we discuss the role of Toll-like receptors (TLRs) as sentinels of uterine immunity in the context of response to pathogens. We highlight the idea that particular TLR activations lead to differential immune cascades that induce preterm birth. Secondly, two alternative routes of pathogenic entry may prove to be critical for inducing preterm birth via a cytokine storm or a secondary and currently unknown cell-mediated mechanism of uterine inflammation. This paper summarizes pathways that underlie activation of adverse and diverse immune responses to foreign agents that may result in preterm birth.


Asunto(s)
Infecciones Bacterianas/inmunología , Nacimiento Prematuro/inmunología , Receptores Toll-Like/inmunología , Virosis/inmunología , Corioamnionitis/inmunología , Corioamnionitis/microbiología , Femenino , Humanos , Trabajo de Parto Prematuro/inmunología , Embarazo , Receptores Toll-Like/fisiología , Útero/inmunología
17.
Cancer Immunol Res ; 8(5): 587-595, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32075802

RESUMEN

The requisites for protein translation in T cells are poorly understood and how translation shapes the antitumor efficacy of T cells is unknown. Here we demonstrated that IL15-conditioned T cells were primed by the metabolic energy sensor AMP-activated protein kinase to undergo diminished translation relative to effector T cells. However, we showed that IL15-conditioned T cells exhibited a remarkable capacity to enhance their protein translation in tumors, which effector T cells were unable to duplicate. Studying the modulation of translation for applications in cancer immunotherapy revealed that direct ex vivo pharmacologic inhibition of translation elongation primed robust T-cell antitumor immunity. Our work elucidates that altering protein translation in CD8+ T cells can shape their antitumor capability.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunoterapia/métodos , Activación de Linfocitos/inmunología , Neoplasias/inmunología , Factor 2 de Elongación Peptídica/metabolismo , Biosíntesis de Proteínas , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Interleucina-15/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias/metabolismo , Neoplasias/terapia
18.
Health Psychol ; 39(9): 745-757, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32833476

RESUMEN

OBJECTIVES: Physiological stress responses have been suggested as a mechanism through which social and biological factors contribute to racial disparities in breast cancer outcomes. Many African Americans experience stressful life events and circumstances. These social factors may contribute to an increased risk of advanced stage disease at diagnosis and/or faster progression, but not all African American women exposed to adverse social factors develop advanced stage disease. Similarly, women who have a limited number of stressors can develop advanced stage breast cancer. Highly individualized stress reactivity may account for these inconsistent associations. METHOD: This report describes the rationale, design, and methods for an exploratory study that uses the experimental medicine approach to: (a) characterize the nature and distribution of stress reactivity among African American breast cancer survivors based on socioeconomic, clinical, and social stressors; (b) examine the impact of stress reactivity on temporal discounting; and (c) determine the extent to which stress reactivity and temporal discounting are associated with adherence to recommendations for cancer control behaviors and treatment compliance as part of the Science of Behavior Change Network. RESULTS: This study addresses several empirical gaps about the most effective ways to develop behavior change interventions for a medically underserved population that continues to experience disparities in cancer morbidity and mortality. CONCLUSIONS: Results from this research will provide the empirical and conceptual basis for future intervention protocols that target mechanisms that are critical to disparities in African American breast cancer survivors. (PsycInfo Database Record (c) 2020 APA, all rights reserved).


Asunto(s)
Supervivientes de Cáncer/psicología , Disparidades en Atención de Salud/normas , Estrés Psicológico/psicología , Adulto , Femenino , Humanos
19.
Cancer Immunol Res ; 7(3): 476-486, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30659052

RESUMEN

Tumor antigen-specific T cells rapidly lose energy and effector function in tumors. The cellular mechanisms by which energy loss and inhibition of effector function occur in tumor-infiltrating lymphocytes (TILs) are ill-defined, and methods to identify tumor antigen-specific TILs that experience such stress are unknown. Processes upstream of the mitochondria guide cell-intrinsic energy depletion. We hypothesized that a mechanism of T-cell-intrinsic energy consumption was the process of oxidative protein folding and disulfide bond formation that takes place in the endoplasmic reticulum (ER) guided by protein kinase R-like endoplasmic reticulum kinase (PERK) and downstream PERK axis target ER oxidoreductase 1 (ERO1α). To test this hypothesis, we created TCR transgenic mice with a T-cell-specific PERK gene deletion (OT1 + Lckcre+ PERK f/f , PERK KO). We found that PERK KO and T cells that were pharmacologically inhibited by PERK or ERO1α maintained reserve energy and exhibited a protein profile consistent with reduced oxidative stress. These T-cell groups displayed superior tumor control compared with T effectors. We identified a biomarker of ER-induced mitochondrial exhaustion in T cells as mitochondrial reactive oxygen species (mtROS), and found that PD-1+ tumor antigen-specific CD8+ TILs express mtROS. In vivo treatment with a PERK inhibitor abrogated mtROS in PD-1+ CD8+ TILs and bolstered CD8+ TIL viability. Combination therapy enabled 100% survival and 71% tumor clearance in a sarcoma mouse model. Our data identify the ER as a regulator of T-cell energetics and indicate that ER elements are effective targets to improve cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Linfocitos Infiltrantes de Tumor/metabolismo , Mitocondrias/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Técnicas de Inactivación de Genes , Humanos , Memoria Inmunológica , Inmunoterapia , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Oxidorreductasas/antagonistas & inhibidores , Oxidorreductasas/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sarcoma/patología , Sarcoma/terapia , Transducción de Señal , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
20.
Cells ; 8(12)2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31779147

RESUMEN

Effective cancer therapies simultaneously restrict tumor cell growth and improve anti-tumor immune responses. Targeting redox-dependent protein folding enzymes within the endoplasmic reticulum (ER) is an alternative approach to activation of the unfolded protein response (UPR) and a novel therapeutic platform to induce malignant cell death. E64FC26 is a recently identified protein disulfide isomerase (PDI) inhibitor that activates the UPR, oxidative stress, and apoptosis in tumor cells, but not normal cell types. Given that targeting cellular redox homeostasis is a strategy to augment T cell tumor control, we tested the effect of E64FC26 on healthy and oncogenic T cells. In stark contrast to the pro-UPR and pro-death effects we observed in malignant T cells, we found that E64FC26 improved viability and limited the UPR in healthy T cells. E64FC26 treatment also diminished oxidative stress and decreased global PDI expression in normal T cells. Oxidative stress and cell death are limited in memory T cells and we found that PDI inhibition promoted memory traits and reshaped T cell metabolism. Using adoptive transfer of tumor antigen-specific CD8 T cells, we demonstrate that T cells activated and expanded in the presence of E64FC26 control tumor growth better than vehicle-matched controls. Our data indicate that PDI inhibitors are a new class of drug that may dually inhibit tumor cell growth and improve T cell tumor control.


Asunto(s)
Estrés del Retículo Endoplásmico , Inmunoterapia Adoptiva , Neoplasias/etiología , Neoplasias/metabolismo , Proteína Disulfuro Isomerasas/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Línea Celular , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Inmunoterapia Adoptiva/métodos , Interleucina-15/metabolismo , Melanoma Experimental , Ratones , Ratones Transgénicos , Neoplasias/patología , Neoplasias/terapia , Oxidación-Reducción , Estrés Oxidativo , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/metabolismo , Respuesta de Proteína Desplegada
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA