Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Cancer Res Commun ; 3(12): 2430-2446, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37971169

RESUMEN

Understanding the intricate dynamics between adoptively transferred immune cells and the brain tumor immune microenvironment (TIME) is crucial for the development of effective T cell-based immunotherapies. In this study, we investigated the influence of the TIME and chimeric antigen receptor (CAR) design on the anti-glioma activity of B7-H3-specific CAR T-cells. Using an immunocompetent glioma model, we evaluated a panel of seven fully murine B7-H3 CARs with variations in transmembrane, costimulatory, and activation domains. We then investigated changes in the TIME following CAR T-cell therapy using high-dimensional flow cytometry and single-cell RNA sequencing. Our results show that five out of six B7-H3 CARs with single costimulatory domains demonstrated robust functionality in vitro. However, these CARs had significantly varied levels of antitumor activity in vivo. To enhance therapeutic effectiveness and persistence, we incorporated 41BB and CD28 costimulation through transgenic expression of 41BBL on CD28-based CAR T-cells. This CAR design was associated with significantly improved anti-glioma efficacy in vitro but did not result in similar improvements in vivo. Analysis of the TIME revealed that CAR T-cell therapy influenced the composition of the TIME, with the recruitment and activation of distinct macrophage and endogenous T-cell subsets crucial for successful antitumor responses. Indeed, complete brain macrophage depletion using a CSF1R inhibitor abrogated CAR T-cell antitumor activity. In sum, our study highlights the critical role of CAR design and its modulation of the TIME in mediating the efficacy of adoptive immunotherapy for high-grade glioma. SIGNIFICANCE: CAR T-cell immunotherapies hold great potential for treating brain cancers; however, they are hindered by a challenging immune environment that dampens their effectiveness. In this study, we show that the CAR design influences the makeup of the immune environment in brain tumors, underscoring the need to target specific immune components to improve CAR T-cell performance, and highlighting the significance of using models with functional immune systems to optimize this therapy.


Asunto(s)
Neoplasias Encefálicas , Glioma , Receptores Quiméricos de Antígenos , Ratones , Animales , Receptores Quiméricos de Antígenos/genética , Linfocitos T , Macrófagos Asociados a Tumores/metabolismo , Antígenos CD28/genética , Glioma/terapia , Neoplasias Encefálicas/terapia , Encéfalo/metabolismo , Microambiente Tumoral
2.
Cell Rep ; 37(11): 110124, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34910919

RESUMEN

Regulatory T (Treg) cells play crucial roles in suppressing deleterious immune response. Here, we investigate how Treg cells are mechanistically induced in vitro (iTreg) and stabilized via transcriptional regulation of Treg lineage-specifying factor Foxp3. We find that acetylation of histone tails at the Foxp3 promoter is required for inducing Foxp3 transcription. Upon induction, histone acetylation signals via bromodomain-containing proteins, particularly targets of inhibitor JQ1, and sustains Foxp3 transcription via a global or trans effect. Subsequently, Tet-mediated DNA demethylation of Foxp3 cis-regulatory elements, mainly enhancer CNS2, increases chromatin accessibility and protein binding, stabilizing Foxp3 transcription and obviating the need for the histone acetylation signal. These processes transform stochastic iTreg induction into a stable cell fate, with the former sensitive and the latter resistant to genetic and environmental perturbations. Thus, sequential histone acetylation and DNA demethylation in Foxp3 induction and maintenance reflect stepwise mechanical switches governing iTreg cell lineage specification.


Asunto(s)
Desmetilación del ADN , Proteínas de Unión al ADN/fisiología , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Histonas/química , Proteínas Proto-Oncogénicas/fisiología , Linfocitos T Reguladores/inmunología , Acetilación , Animales , Diferenciación Celular , Metilación de ADN , Femenino , Factores de Transcripción Forkhead/genética , Histonas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos
3.
J Invest Dermatol ; 139(1): 62-70, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30030152

RESUMEN

High-risk skin cancer is a rare, but severe, complication associated with discoid lupus erythematosus (DLE). Chronic scar, inflammation, UVR, and immunosuppressive medications are proposed explanations for this heightened skin cancer risk; however, the exact mechanism driving skin carcinogenesis in DLE is unknown. The distinct co-localization of multiple independent skin cancers with areas of active inflammation in two DLE patients followed over 8 years strongly suggested that lupus inflammation promotes skin carcinogenesis in DLE. To investigate this clinical observation, we subjected lupus-prone MRL/lpr and control (MRL/n) mice to a skin carcinogenesis protocol. Skin tumors developed preferentially within the cutaneous lupus inflammation without scarring in MRL/lpr mice (P < 0.01). The inflammation in MRL/lpr skin was characterized by the accumulation of regulatory T cells, mast cells, M2 macrophages, and markedly elevated transforming growth factor-ß1 and IL-6 levels, which have been linked to tumor promotion. Tacrolimus treatment reduced skin inflammation and blocked cancer development in MRL/lpr mice (P = 0.0195). A similar tumor-promoting immune environment was detected in SCCs and the perilesional skin of cancer-prone DLE patients. Therefore, discoid lupus inflammation promotes skin cancer in high-risk DLE patients, and blocking the inflammation may be critical for preventing this life-threatening complication of DLE.


Asunto(s)
Citocinas/metabolismo , Inflamación/patología , Lupus Eritematoso Discoide/patología , Neoplasias Cutáneas/etiología , Piel/patología , Animales , Carcinogénesis , Enfermedad Crónica , Femenino , Humanos , Inflamación/complicaciones , Inflamación/metabolismo , Lupus Eritematoso Discoide/complicaciones , Lupus Eritematoso Discoide/metabolismo , Ratones , Ratones Endogámicos MRL lpr , Persona de Mediana Edad , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
4.
Sci Rep ; 7(1): 12751, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28986576

RESUMEN

Currently ablative fractional photothermolysis (aFP) with CO2 laser is used for a wide variety of dermatological indications. This study presents and discusses the utility of aFP for treating oncological indications. We used a fractional CO2 laser and anti-PD-1 inhibitor to treat a tumor established unilaterally by the CT26 wild type (CT26WT) colon carcinoma cell line. Inoculated tumors grew significantly slower in aFP-treated groups (aFP and aFP + anti-PD-1 groups) and complete remission was observed in the aFP-treated groups. Flow cytometric analysis showed aFP treatment elicited an increase of CD3+, CD4+, CD8+ vand epitope specific CD8+ T cells. Moreover, the ratio of CD8+ T cells to Treg increased in the aFP-treated groups. Additionally, we established a bilateral CT26WT-inoculated mouse model, treating tumors on one-side and observing both tumors. Interestingly, tumors grew significantly slower in the aFP + anti-PD-1 groups and complete remission was observed for tumors on both aFP-treated and untreated sides. This study has demonstrated a potential role of aFP treatments in oncology.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Inmunidad , Láseres de Gas , Neoplasias/inmunología , Inmunidad Adaptativa , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Epítopos/metabolismo , Femenino , Terapia por Láser , Recuento de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos BALB C , Fotólisis , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Análisis de Supervivencia , Carga Tumoral
5.
J Clin Invest ; 127(1): 106-116, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27869649

RESUMEN

BACKGROUND: Actinic keratosis is a precursor to cutaneous squamous cell carcinoma. Long treatment durations and severe side effects have limited the efficacy of current actinic keratosis treatments. Thymic stromal lymphopoietin (TSLP) is an epithelium-derived cytokine that induces a robust antitumor immunity in barrier-defective skin. Here, we investigated the efficacy of calcipotriol, a topical TSLP inducer, in combination with 5-fluorouracil (5-FU) as an immunotherapy for actinic keratosis. METHODS: The mechanism of calcipotriol action against skin carcinogenesis was examined in genetically engineered mouse models. The efficacy and safety of 0.005% calcipotriol ointment combined with 5% 5-FU cream were compared with Vaseline plus 5-FU for the field treatment of actinic keratosis in a randomized, double-blind clinical trial involving 131 participants. The assigned treatment was self-applied to the entirety of the qualified anatomical sites (face, scalp, and upper extremities) twice daily for 4 consecutive days. The percentage of reduction in the number of actinic keratoses (primary outcome), local skin reactions, and immune activation parameters were assessed. RESULTS: Calcipotriol suppressed skin cancer development in mice in a TSLP-dependent manner. Four-day application of calcipotriol plus 5-FU versus Vaseline plus 5-FU led to an 87.8% versus 26.3% mean reduction in the number of actinic keratoses in participants (P < 0.0001). Importantly, calcipotriol plus 5-FU treatment induced TSLP, HLA class II, and natural killer cell group 2D (NKG2D) ligand expression in the lesional keratinocytes associated with a marked CD4+ T cell infiltration, which peaked on days 10-11 after treatment, without pain, crusting, or ulceration. CONCLUSION: Our findings demonstrate the synergistic effects of calcipotriol and 5-FU treatment in optimally activating a CD4+ T cell-mediated immunity against actinic keratoses and, potentially, cancers of the skin and other organs. TRIAL REGISTRATION: ClinicalTrials.gov NCT02019355. FUNDING: Not applicable (investigator-initiated clinical trial).


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma de Células Escamosas/tratamiento farmacológico , Queratosis Actínica/tratamiento farmacológico , Lesiones Precancerosas/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Administración Tópica , Anciano , Anciano de 80 o más Años , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Calcitriol/administración & dosificación , Calcitriol/análogos & derivados , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Citocinas/genética , Citocinas/inmunología , Femenino , Fluorouracilo/administración & dosificación , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Queratosis Actínica/genética , Queratosis Actínica/inmunología , Queratosis Actínica/patología , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Lesiones Precancerosas/genética , Lesiones Precancerosas/inmunología , Lesiones Precancerosas/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Linfopoyetina del Estroma Tímico
6.
J Clin Invest ; 126(4): 1458-70, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26927668

RESUMEN

Advances in the field of cancer immunology, including studies on tumor-infiltrating CD8+ cytotoxic T lymphocytes (CTLs), have led to new immunotherapeutics with proven efficacy against late-stage cancers. However, the antitumor potential of the immune system in targeting early-stage cancers remains uncertain. Here, we demonstrated that both genetic and chemical induction of thymic stromal lymphopoietin (TSLP) at a distant site leads to robust antitumor immunity against spontaneous breast carcinogenesis in mice. Breast tumors exposed to high circulating levels of TSLP were arrested at an early adenoma-like stage and were prevented from advancing to late carcinoma and metastasis. Additionally, CD4+ Th2 cells mediated the antitumor effects of TSLP, challenging the notion that Th2 cells only promote cancer. We also discovered that TSLP is expressed by the breast tumor cells themselves and acts to block breast cancer promotion. Moreover, TSLP-induced immunity also blocked early stages of pancreatic cancer development. Together, our findings demonstrate that TSLP potently induces immunity directed against early stages of breast cancer development without causing inflammation in the normal breast tissue. Moreover, our results highlight a previously unappreciated function of the immune system in controlling the early development of cancer and establish a fundamental role for TSLP and Th2 cells in tumor immunity against early-stage cancers.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Inmunidad Celular , Neoplasias Mamarias Experimentales/inmunología , Células Th2/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Linfocitos T CD8-positivos/patología , Citocinas/genética , Femenino , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Células Th2/patología , Proteínas Supresoras de Tumor/genética , Linfopoyetina del Estroma Tímico
7.
J Clin Invest ; 124(11): 5037-41, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25295539

RESUMEN

Allergic contact dermatitis (ACD) is well recognized as an adverse event associated with implantable medical devices that contain allergenic materials like nickel; however, other cutaneous consequences of chronic exposure to allergens in implanted devices are not well understood. Here, we present a clinical case of Marjolin's ulcer, an invasive squamous cell carcinoma (SCC) that developed in response to chronic ACD caused by an orthopedic implant. We used a standard murine model of contact hypersensitivity to determine whether chronic ACD promotes skin carcinogenesis. Chronic application of 1-fluoro-2,4-dinitrobenzene (DNFB) to carcinogen-treated skin led to the development of papillomas and aggressive SCC. DNFB-driven chronic ACD was marked by type 2 inflammation, which mediated skin carcinogenesis, as mice unable to mount an inflammatory response were less likely to develop skin tumors. Importantly, we found similar tumor-promoting inflammation surrounding the SCC in our patient. Our findings demonstrate that chronic ACD caused by constant exposure to an allergen can promote tumorigenesis at skin sites with preexisting cancer-initiated cells. Moreover, our results suggest that patients with implantable devices placed in close proximity to the skin should be monitored for ACD and highlight the importance of patch testing prior to the placement of such devices.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico , Dermatitis Alérgica por Contacto/complicaciones , Neoplasias Cutáneas/diagnóstico , Animales , Carcinogénesis/inmunología , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/cirugía , Enfermedad Crónica , Dermatitis Alérgica por Contacto/patología , Femenino , Humanos , Fijadores Internos/efectos adversos , Ratones Endogámicos C57BL , Persona de Mediana Edad , Níquel/inmunología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA