Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38464258

RESUMEN

The modern armamentarium for cancer treatment includes immunotherapy and targeted therapy, such as protein kinase inhibitors. However, the mechanisms that allow cancer-targeting drugs to effectively mobilize dendritic cells (DCs) and affect immunotherapy are poorly understood. Here, we report that among shared gene targets of clinically relevant protein kinase inhibitors, high PIKFYVE expression was least predictive of complete response in patients who received immune checkpoint blockade (ICB). In immune cells, high PIKFYVE expression in DCs was associated with worse response to ICB. Genetic and pharmacological studies demonstrated that PIKfyve ablation enhanced DC function via selectively altering the alternate/non-canonical NF-κB pathway. Both loss of Pikfyve in DCs and treatment with apilimod, a potent and specific PIKfyve inhibitor, restrained tumor growth, enhanced DC-dependent T cell immunity, and potentiated ICB efficacy in tumor-bearing mouse models. Furthermore, the combination of a vaccine adjuvant and apilimod reduced tumor progression in vivo. Thus, PIKfyve negatively controls DCs, and PIKfyve inhibition has promise for cancer immunotherapy and vaccine treatment strategies.

2.
Cell Rep ; 43(3): 113942, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38489266

RESUMEN

Tumor-associated macrophages (TAMs) shape tumor immunity and therapeutic efficacy. However, it is poorly understood whether and how post-translational modifications (PTMs) intrinsically affect the phenotype and function of TAMs. Here, we reveal that peptidylarginine deiminase 4 (PAD4) exhibits the highest expression among common PTM enzymes in TAMs and negatively correlates with the clinical response to immune checkpoint blockade. Genetic and pharmacological inhibition of PAD4 in macrophages prevents tumor progression in tumor-bearing mouse models, accompanied by an increase in macrophage major histocompatibility complex (MHC) class II expression and T cell effector function. Mechanistically, PAD4 citrullinates STAT1 at arginine 121, thereby promoting the interaction between STAT1 and protein inhibitor of activated STAT1 (PIAS1), and the loss of PAD4 abolishes this interaction, ablating the inhibitory role of PIAS1 in the expression of MHC class II machinery in macrophages and enhancing T cell activation. Thus, the PAD4-STAT1-PIAS1 axis is an immune restriction mechanism in macrophages and may serve as a cancer immunotherapy target.


Asunto(s)
Hidrolasas , Procesamiento Proteico-Postraduccional , Ratones , Animales , Desiminasas de la Arginina Proteica/metabolismo , Arginina Deiminasa Proteína-Tipo 4/genética , Arginina Deiminasa Proteína-Tipo 4/metabolismo , Hidrolasas/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Macrófagos/metabolismo
3.
Science ; 383(6678): 62-70, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38175892

RESUMEN

Immune checkpoint inhibitors can stimulate antitumor immunity but can also induce toxicities termed immune-related adverse events (irAEs). Colitis is a common and severe irAE that can lead to treatment discontinuation. Mechanistic understanding of gut irAEs has been hampered because robust colitis is not observed in laboratory mice treated with checkpoint inhibitors. We report here that this limitation can be overcome by using mice harboring the microbiota of wild-caught mice, which develop overt colitis following treatment with anti-CTLA-4 antibodies. Intestinal inflammation is driven by unrestrained activation of IFNγ-producing CD4+ T cells and depletion of peripherally induced regulatory T cells through Fcγ receptor signaling. Accordingly, anti-CTLA-4 nanobodies that lack an Fc domain can promote antitumor responses without triggering colitis. This work suggests a strategy for mitigating gut irAEs while preserving antitumor stimulating effects of CTLA-4 blockade.


Asunto(s)
Linfocitos T CD4-Positivos , Colitis , Inhibidores de Puntos de Control Inmunológico , Activación de Linfocitos , Microbiota , Receptores de IgG , Animales , Ratones , Linfocitos T CD4-Positivos/inmunología , Colitis/etiología , Colitis/microbiología , Antígeno CTLA-4/antagonistas & inhibidores , Microbiota/inmunología , Receptores de IgG/inmunología , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Ratones Endogámicos C57BL
4.
Cancer Cell ; 41(2): 304-322.e7, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36638784

RESUMEN

Immune checkpoint blockade (ICB) can produce durable responses against cancer. We and others have found that a subset of patients experiences paradoxical rapid cancer progression during immunotherapy. It is poorly understood how tumors can accelerate their progression during ICB. In some preclinical models, ICB causes hyperprogressive disease (HPD). While immune exclusion drives resistance to ICB, counterintuitively, patients with HPD and complete response (CR) following ICB manifest comparable levels of tumor-infiltrating CD8+ T cells and interferon γ (IFNγ) gene signature. Interestingly, patients with HPD but not CR exhibit elevated tumoral fibroblast growth factor 2 (FGF2) and ß-catenin signaling. In animal models, T cell-derived IFNγ promotes tumor FGF2 signaling, thereby suppressing PKM2 activity and decreasing NAD+, resulting in reduction of SIRT1-mediated ß-catenin deacetylation and enhanced ß-catenin acetylation, consequently reprograming tumor stemness. Targeting the IFNγ-PKM2-ß-catenin axis prevents HPD in preclinical models. Thus, the crosstalk of core immunogenic, metabolic, and oncogenic pathways via the IFNγ-PKM2-ß-catenin cascade underlies ICB-associated HPD.


Asunto(s)
Neoplasias , beta Catenina , Animales , Linfocitos T CD8-positivos , Factor 2 de Crecimiento de Fibroblastos , Neoplasias/terapia , Neoplasias/patología , Progresión de la Enfermedad , Interferón gamma , Inmunoterapia/métodos
5.
Cell Rep ; 39(1): 110609, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35385733

RESUMEN

Tumor-associated macrophages (TAMs) are a major cellular component in the tumor microenvironment (TME). However, the relationship between the phenotype and metabolic pattern of TAMs remains poorly understood. We performed single-cell transcriptome profiling on hepatic TAMs from mice bearing liver metastatic tumors. We find that TAMs manifest high heterogeneity at the levels of transcription, development, metabolism, and function. Integrative analyses and validation experiments indicate that increased purine metabolism is a feature of TAMs with pro-tumor and terminal differentiation phenotypes. Like mouse TAMs, human TAMs are highly heterogeneous. Human TAMs with increased purine metabolism exhibit a pro-tumor phenotype and correlate with poor therapeutic efficacy to immune checkpoint blockade. Altogether, our work demonstrates that TAMs are developmentally, metabolically, and functionally heterogeneous and purine metabolism may be a key metabolic feature of a pro-tumor macrophage population.


Asunto(s)
Neoplasias Hepáticas , Microambiente Tumoral , Animales , Perfilación de la Expresión Génica , Neoplasias Hepáticas/patología , Macrófagos/metabolismo , Ratones , Macrófagos Asociados a Tumores
6.
Oncoimmunology ; 11(1): 2052640, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309733

RESUMEN

Chronic inflammation and oncogenic pathway activation are key-contributing factors in colorectal cancer pathogenesis. However, colorectal intrinsic mechanisms linking these two factors in cancer development are poorly defined. Here, we show that intestinal epithelial cell (IEC)-specific deletion of Dot1l histone methyltransferase (Dot1lΔIEC ) reduced H3K79 dimethylation (H3K79me2) in IECs and inhibited intestinal tumor formation in ApcMin - and AOM-DSS-induced colorectal cancer models. IEC-Dot1l abrogation was accompanied by alleviative colorectal inflammation and reduced Wnt/ß-catenin signaling activation. Mechanistically, Dot1l deficiency resulted in an increase in Foxp3+RORϒ+ regulatory T (Treg) cells and a decrease in inflammatory Th17 and Th22 cells, thereby reducing local inflammation in the intestinal tumor microenvironment. Furthermore, Dot1l deficiency caused a reduction of H3K79me2 occupancies in the promoters of the Wnt/ß-catenin signaling genes, thereby diminishing Wnt/ß-catenin oncogenic signaling pathway activation in colorectal cancer cells. Clinically, high levels of tumor H3K79me2 were detected in patients with colorectal carcinomas as compared to adenomas, and negatively correlated with RORϒ+FOXP3+ Treg cells. Altogether, we conclude that DOT1L is an intrinsic molecular node connecting chronic immune activation and oncogenic signaling pathways in colorectal cancer. Our work suggests that targeting the DOT1L pathway may control colorectal carcinogenesis. Significance: IEC-intrinsic DOT1L controls T cell subset balance and key oncogenic pathway activation, impacting colorectal carcinogenesis.


Asunto(s)
Neoplasias Colorrectales , N-Metiltransferasa de Histona-Lisina , Subgrupos de Linfocitos T , Carcinogénesis/metabolismo , Neoplasias Colorrectales/patología , Factores de Transcripción Forkhead/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Inflamación , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Microambiente Tumoral , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
7.
Nat Cell Biol ; 23(5): 526-537, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33958760

RESUMEN

Major histocompatibility complex-I (MHC-I) presents tumour antigens to CD8+ T cells and triggers anti-tumour immunity. Humans may have 30,000-60,000 long noncoding RNAs (lncRNAs). However, it remains poorly understood whether lncRNAs affect tumour immunity. Here, we identify a lncRNA, lncRNA inducing MHC-I and immunogenicity of tumour (LIMIT), in humans and mice. We found that IFNγ stimulated LIMIT, LIMIT cis-activated the guanylate-binding protein (GBP) gene cluster and GBPs disrupted the association between HSP90 and heat shock factor-1 (HSF1), thereby resulting in HSF1 activation and transcription of MHC-I machinery, but not PD-L1. RNA-guided CRISPR activation of LIMIT boosted GBPs and MHC-I, and potentiated tumour immunogenicity and checkpoint therapy. Silencing LIMIT, GBPs and/or HSF1 diminished MHC-I, impaired antitumour immunity and blunted immunotherapy efficacy. Clinically, LIMIT, GBP- and HSF1-signalling transcripts and proteins correlated with MHC-I, tumour-infiltrating T cells and checkpoint blockade response in patients with cancer. Together, we demonstrate that LIMIT is a cancer immunogenic lncRNA and the LIMIT-GBP-HSF1 axis may be targetable for cancer immunotherapy.


Asunto(s)
Inmunoterapia , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , ARN Largo no Codificante/genética , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Inmunoterapia/métodos , Neoplasias/inmunología , Transducción de Señal/fisiología
8.
Nat Immunol ; 22(4): 460-470, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33767425

RESUMEN

Targeting the p53-MDM2 pathway to reactivate tumor p53 is a chemotherapeutic approach. However, the involvement of this pathway in CD8+ T cell-mediated antitumor immunity is unknown. Here, we report that mice with MDM2 deficiency in T cells exhibit accelerated tumor progression and a decrease in tumor-infiltrating CD8+ T cell survival and function. Mechanistically, MDM2 competes with c-Cbl for STAT5 binding, reduces c-Cbl-mediated STAT5 degradation and enhances STAT5 stability in tumor-infiltrating CD8+ T cells. Targeting the p53-MDM2 interaction with a pharmacological agent, APG-115, augmented MDM2 in T cells, thereby stabilizing STAT5, boosting T cell immunity and synergizing with cancer immunotherapy. Unexpectedly, these effects of APG-115 were dependent on p53 and MDM2 in T cells. Clinically, MDM2 abundance correlated with T cell function and interferon-γ signature in patients with cancer. Thus, the p53-MDM2 pathway controls T cell immunity, and targeting this pathway may treat patients with cancer regardless of tumor p53 status.


Asunto(s)
Linfocitos T CD8-positivos/enzimología , Linfocitos Infiltrantes de Tumor/enzimología , Neoplasias/enzimología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Antineoplásicos/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Línea Celular Tumoral , Terapia Combinada , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Inmunoterapia Adoptiva , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/trasplante , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Estabilidad Proteica , Proteolisis , Proteínas Proto-Oncogénicas c-mdm2/genética , Factor de Transcripción STAT5/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
9.
Cancer Discov ; 11(7): 1826-1843, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33627378

RESUMEN

Mutations in IFN and MHC signaling genes endow immunotherapy resistance. Patients with colorectal cancer infrequently exhibit IFN and MHC signaling gene mutations and are generally resistant to immunotherapy. In exploring the integrity of IFN and MHC signaling in colorectal cancer, we found that optineurin was a shared node between the two pathways and predicted colorectal cancer patient outcome. Loss of optineurin occurs in early-stage human colorectal cancer. Immunologically, optineurin deficiency was shown to attenuate IFNGR1 and MHC-I expression, impair T-cell immunity, and diminish immunotherapy efficacy in murine cancer models and patients with cancer. Mechanistically, we observed that IFNGR1 was S-palmitoylated on Cys122, and AP3D1 bound with and sorted palmitoylated IFNGR1 to lysosome for degradation. Unexpectedly, optineurin interacted with AP3D1 to prevent palmitoylated IFNGR1 lysosomal sorting and degradation, thereby maintaining IFNγ and MHC-I signaling integrity. Furthermore, pharmacologically targeting IFNGR1 palmitoylation stabilized IFNGR1, augmented tumor immunity, and sensitized checkpoint therapy. Thus, loss of optineurin drives immune evasion and intrinsic immunotherapy resistance in colorectal cancer. SIGNIFICANCE: Loss of optineurin impairs the integrity of both IFNγ and MHC-I signaling pathways via palmitoylation-dependent IFNGR1 lysosomal sorting and degradation, thereby driving immune evasion and intrinsic immunotherapy resistance in colorectal cancer. Our work suggests that pharmacologically targeting IFNGR1 palmitoylation can stabilize IFNGR1, enhance T-cell immunity, and sensitize checkpoint therapy in colorectal cancer.See related commentary by Salvagno and Cubillos-Ruiz, p. 1623.This article is highlighted in the In This Issue feature, p. 1601.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorrectales/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Receptores de Interferón/metabolismo , Animales , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Femenino , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Interferón gamma/metabolismo , Lipoilación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Transporte de Proteínas , Organismos Libres de Patógenos Específicos , Receptor de Interferón gamma
10.
Nat Med ; 27(1): 152-164, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33398162

RESUMEN

Metastasis is the primary cause of cancer mortality, and cancer frequently metastasizes to the liver. It is not clear whether liver immune tolerance mechanisms contribute to cancer outcomes. We report that liver metastases diminish immunotherapy efficacy systemically in patients and preclinical models. Patients with liver metastases derive limited benefit from immunotherapy independent of other established biomarkers of response. In multiple mouse models, we show that liver metastases siphon activated CD8+ T cells from systemic circulation. Within the liver, activated antigen-specific Fas+CD8+ T cells undergo apoptosis following their interaction with FasL+CD11b+F4/80+ monocyte-derived macrophages. Consequently, liver metastases create a systemic immune desert in preclinical models. Similarly, patients with liver metastases have reduced peripheral T cell numbers and diminished tumoral T cell diversity and function. In preclinical models, liver-directed radiotherapy eliminates immunosuppressive hepatic macrophages, increases hepatic T cell survival and reduces hepatic siphoning of T cells. Thus, liver metastases co-opt host peripheral tolerance mechanisms to cause acquired immunotherapy resistance through CD8+ T cell deletion, and the combination of liver-directed radiotherapy and immunotherapy could promote systemic antitumor immunity.


Asunto(s)
Inmunoterapia , Neoplasias Hepáticas Experimentales/secundario , Neoplasias Hepáticas Experimentales/terapia , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Macrófagos/inmunología , Linfocitos T/inmunología , Animales , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/secundario , Carcinoma de Pulmón de Células no Pequeñas/terapia , Línea Celular Tumoral , Estudios de Cohortes , Terapia Combinada , Femenino , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas Experimentales/inmunología , Activación de Linfocitos , Masculino , Melanoma/inmunología , Melanoma/secundario , Melanoma/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Radioterapia Adyuvante , Linfocitos T/clasificación , Linfocitos T/patología , Insuficiencia del Tratamiento , Resultado del Tratamiento , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación
11.
Cancer Cell ; 39(4): 480-493.e6, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33513345

RESUMEN

Immunotherapy induces durable clinical responses in a fraction of patients with cancer. However, therapeutic resistance poses a major challenge to current immunotherapies. Here, we identify that expression of tumor stanniocalcin 1 (STC1) correlates with immunotherapy efficacy and is negatively associated with patient survival across diverse cancer types. Gain- and loss-of-function experiments demonstrate that tumor STC1 supports tumor progression and enables tumor resistance to checkpoint blockade in murine tumor models. Mechanistically, tumor STC1 interacts with calreticulin (CRT), an "eat-me" signal, and minimizes CRT membrane exposure, thereby abrogating membrane CRT-directed phagocytosis by antigen-presenting cells (APCs), including macrophages and dendritic cells. Consequently, this impairs APC capacity of antigen presentation and T cell activation. Thus, tumor STC1 inhibits APC phagocytosis and contributes to tumor immune evasion and immunotherapy resistance. We suggest that STC1 is a previously unappreciated phagocytosis checkpoint and targeting STC1 and its interaction with CRT may sensitize to cancer immunotherapy.


Asunto(s)
Glicoproteínas/metabolismo , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Fagocitosis/inmunología , Escape del Tumor/inmunología , Animales , Presentación de Antígeno/inmunología , Inmunoterapia/métodos , Macrófagos/metabolismo , Ratones , Fagocitosis/efectos de los fármacos , Receptores Inmunológicos/inmunología
12.
Nature ; 585(7824): 277-282, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32879489

RESUMEN

Abnormal epigenetic patterns correlate with effector T cell malfunction in tumours1-4, but the cause of this link is unknown. Here we show that tumour cells disrupt methionine metabolism in CD8+ T cells, thereby lowering intracellular levels of methionine and the methyl donor S-adenosylmethionine (SAM) and resulting in loss of dimethylation at lysine 79 of histone H3 (H3K79me2). Loss of H3K79me2 led to low expression of STAT5 and impaired T cell immunity. Mechanistically, tumour cells avidly consumed methionine and outcompeted T cells for methionine by expressing high levels of the methionine transporter SLC43A2. Genetic and biochemical inhibition of tumour SLC43A2 restored H3K79me2 in T cells, thereby boosting spontaneous and checkpoint-induced tumour immunity. Moreover, methionine supplementation improved the expression of H3K79me2 and STAT5 in T cells, and this was accompanied by increased T cell immunity in tumour-bearing mice and patients with colon cancer. Clinically, tumour SLC43A2 correlated negatively with T cell histone methylation and functional gene signatures. Our results identify a mechanistic connection between methionine metabolism, histone patterns, and T cell immunity in the tumour microenvironment. Thus, cancer methionine consumption is an immune evasion mechanism, and targeting cancer methionine signalling may provide an immunotherapeutic approach.


Asunto(s)
Sistema de Transporte de Aminoácidos L/metabolismo , Linfocitos T CD8-positivos/metabolismo , Histonas/metabolismo , Metionina/metabolismo , Metilación , Neoplasias/metabolismo , Sistema de Transporte de Aminoácidos L/deficiencia , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Epigénesis Genética , Femenino , Histonas/química , Humanos , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Factor de Transcripción STAT5/metabolismo
13.
JCI Insight ; 5(18)2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32780724

RESUMEN

Tumor-associated macrophages (TAMs) affect cancer progression and therapy. Ovarian carcinoma often metastasizes to the peritoneal cavity. Here, we found 2 peritoneal macrophage subsets in mice bearing ID8 ovarian cancer based on T cell immunoglobulin and mucin domain containing 4 (Tim-4) expression. Tim-4+ TAMs were embryonically originated and locally sustained while Tim-4- TAMs were replenished from circulating monocytes. Tim-4+ TAMs, but not Tim-4- TAMs, promoted tumor growth in vivo. Relative to Tim-4- TAMs, Tim-4+ TAMs manifested high oxidative phosphorylation and adapted mitophagy to alleviate oxidative stress. High levels of arginase-1 in Tim-4+ TAMs contributed to potent mitophagy activities via weakened mTORC1 activation due to low arginine resultant from arginase-1-mediated metabolism. Furthermore, genetic deficiency of autophagy element FAK family-interacting protein of 200 kDa resulted in Tim-4+ TAM loss via ROS-mediated apoptosis and elevated T cell immunity and ID8 tumor inhibition in vivo. Moreover, human ovarian cancer-associated macrophages positive for complement receptor of the immunoglobulin superfamily (CRIg) were transcriptionally, metabolically, and functionally similar to murine Tim-4+ TAMs. Thus, targeting CRIg+ (Tim-4+) TAMs may potentially treat patients with ovarian cancer with peritoneal metastasis.


Asunto(s)
Autofagia , Macrófagos Peritoneales/patología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Neoplasias Ováricas/patología , Estrés Oxidativo , Neoplasias Peritoneales/secundario , Adaptación Fisiológica , Animales , Proteínas Relacionadas con la Autofagia/fisiología , Femenino , Humanos , Antígenos Comunes de Leucocito/fisiología , Macrófagos Peritoneales/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Ováricas/metabolismo , Neoplasias Peritoneales/metabolismo , Receptores CCR2/fisiología
14.
J Clin Invest ; 130(5): 2712-2726, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32027624

RESUMEN

Whether mutations in cancer driver genes directly affect cancer immune phenotype and T cell immunity remains a standing question. ARID1A is a core member of the polymorphic BRG/BRM-associated factor chromatin remodeling complex. ARID1A mutations occur in human cancers and drive cancer development. Here, we studied the molecular, cellular, and clinical impact of ARID1A aberrations on cancer immunity. We demonstrated that ARID1A aberrations resulted in limited chromatin accessibility to IFN-responsive genes, impaired IFN gene expression, anemic T cell tumor infiltration, poor tumor immunity, and shortened host survival in many human cancer histologies and in murine cancer models. Impaired IFN signaling was associated with poor immunotherapy response. Mechanistically, ARID1A interacted with EZH2 via its carboxyl terminal and antagonized EZH2-mediated IFN responsiveness. Thus, the interaction between ARID1A and EZH2 defines cancer IFN responsiveness and immune evasion. Our work indicates that cancer epigenetic driver mutations can shape cancer immune phenotype and immunotherapy.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Mutación , Neoplasias/genética , Neoplasias/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Animales , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/inmunología , Proteínas de Unión al ADN/química , Proteína Potenciadora del Homólogo Zeste 2/química , Proteína Potenciadora del Homólogo Zeste 2/inmunología , Epigénesis Genética , Femenino , Humanos , Inmunofenotipificación , Inmunoterapia , Interferones/genética , Interferones/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Ratones , Neoplasias/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Transducción de Señal/genética , Transducción de Señal/inmunología , Factores de Transcripción/química , Escape del Tumor/genética , Escape del Tumor/inmunología
15.
Cancer Discov ; 9(12): 1673-1685, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31554642

RESUMEN

A challenge in oncology is to rationally and effectively integrate immunotherapy with traditional modalities, including radiotherapy. Here, we demonstrate that radiotherapy induces tumor-cell ferroptosis. Ferroptosis agonists augment and ferroptosis antagonists limit radiotherapy efficacy in tumor models. Immunotherapy sensitizes tumors to radiotherapy by promoting tumor-cell ferroptosis. Mechanistically, IFNγ derived from immunotherapy-activated CD8+ T cells and radiotherapy-activated ATM independently, yet synergistically, suppresses SLC7A11, a unit of the glutamate-cystine antiporter xc-, resulting in reduced cystine uptake, enhanced tumor lipid oxidation and ferroptosis, and improved tumor control. Thus, ferroptosis is an unappreciated mechanism and focus for the development of effective combinatorial cancer therapy. SIGNIFICANCE: This article describes ferroptosis as a previously unappreciated mechanism of action for radiotherapy. Further, it shows that ferroptosis is a novel point of synergy between immunotherapy and radiotherapy. Finally, it nominates SLC7A11, a critical regulator of ferroptosis, as a mechanistic determinant of synergy between radiotherapy and immunotherapy.This article is highlighted in the In This Issue feature, p. 1631.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/genética , Antineoplásicos Inmunológicos/administración & dosificación , Melanoma Experimental/terapia , Sulfasalazina/administración & dosificación , Animales , Antineoplásicos Inmunológicos/farmacología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Regulación hacia Abajo , Ferroptosis/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoterapia/métodos , Interferón gamma , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/efectos de la radiación , Melanoma Experimental/genética , Ratones , Oxidación-Reducción , Sulfasalazina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Nature ; 569(7755): 270-274, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31043744

RESUMEN

Cancer immunotherapy restores or enhances the effector function of CD8+ T cells in the tumour microenvironment1,2. CD8+ T cells activated by cancer immunotherapy clear tumours mainly by inducing cell death through perforin-granzyme and Fas-Fas ligand pathways3,4. Ferroptosis is a form of cell death that differs from apoptosis and results from iron-dependent accumulation of lipid peroxide5,6. Although it has been investigated in vitro7,8, there is emerging evidence that ferroptosis might be implicated in a variety of pathological scenarios9,10. It is unclear whether, and how, ferroptosis is involved in T cell immunity and cancer immunotherapy. Here we show that immunotherapy-activated CD8+ T cells enhance ferroptosis-specific lipid peroxidation in tumour cells, and that increased ferroptosis contributes to the anti-tumour efficacy of immunotherapy. Mechanistically, interferon gamma (IFNγ) released from CD8+ T cells downregulates the expression of SLC3A2 and SLC7A11, two subunits of the glutamate-cystine antiporter system xc-, impairs the uptake of cystine by tumour cells, and as a consequence, promotes tumour cell lipid peroxidation and ferroptosis. In mouse models, depletion of cystine or cysteine by cyst(e)inase (an engineered enzyme that degrades both cystine and cysteine) in combination with checkpoint blockade synergistically enhanced T cell-mediated anti-tumour immunity and induced ferroptosis in tumour cells. Expression of system xc- was negatively associated, in cancer patients, with CD8+ T cell signature, IFNγ expression, and patient outcome. Analyses of human transcriptomes before and during nivolumab therapy revealed that clinical benefits correlate with reduced expression of SLC3A2 and increased IFNγ and CD8. Thus, T cell-promoted tumour ferroptosis is an anti-tumour mechanism, and targeting this pathway in combination with checkpoint blockade is a potential therapeutic approach.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ferroptosis , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Antígeno B7-H1/antagonistas & inhibidores , Línea Celular Tumoral , Cisteína/metabolismo , Femenino , Ferroptosis/efectos de los fármacos , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Humanos , Interferón gamma/inmunología , Peroxidación de Lípido , Melanoma/genética , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/terapia , Ratones , Neoplasias/metabolismo , Nivolumab/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Resultado del Tratamiento
17.
J Immunol ; 201(2): 814-820, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29802127

RESUMEN

Naive T cells are thought to be functionally quiescent. In this study, we studied and compared the phenotype, cytokine profile, and potential function of human naive CD4+ T cells in umbilical cord and peripheral blood. We found that naive CD4+ T cells, but not memory T cells, expressed high levels of chemokine CXCL8. CXCL8+ naive T cells were preferentially enriched CD31+ T cells and did not express T cell activation markers or typical Th effector cytokines, including IFN-γ, IL-4, IL-17, and IL-22. In addition, upon activation, naive T cells retained high levels of CXCL8 expression. Furthermore, we showed that naive T cell-derived CXCL8 mediated neutrophil migration in the in vitro migration assay, supported tumor sphere formation, and promoted tumor growth in an in vivo human xenograft model. Thus, human naive T cells are phenotypically and functionally heterogeneous and can carry out active functions in immune responses.


Asunto(s)
Células Sanguíneas/fisiología , Interleucina-8/metabolismo , Neoplasias Experimentales/inmunología , Neutrófilos/fisiología , Linfocitos T/fisiología , Cordón Umbilical/patología , Animales , Carcinogénesis , Línea Celular Tumoral , Movimiento Celular , Citocinas/metabolismo , Regulación de la Expresión Génica , Humanos , Interleucina-8/genética , Activación de Linfocitos , Ratones , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cell Metab ; 28(1): 87-103.e6, 2018 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-29805099

RESUMEN

Myeloid-derived suppressor cells (MDSCs) inhibit anti-tumor immunity. Aerobic glycolysis is a hallmark of cancer. However, the link between MDSCs and glycolysis is unknown in patients with triple-negative breast cancer (TNBC). Here, we detect abundant glycolytic activities in human TNBC. In two TNBC mouse models, 4T1 and Py8119, glycolysis restriction inhibits tumor granulocyte colony-stimulating factor (G-CSF) and granulocyte macrophage colony-stimulating factor (GM-CSF) expression and reduces MDSCs. These are accompanied with enhanced T cell immunity, reduced tumor growth and metastasis, and prolonged mouse survival. Mechanistically, glycolysis restriction represses the expression of a specific CCAAT/enhancer-binding protein beta (CEBPB) isoform, liver-enriched activator protein (LAP), via the AMP-activated protein kinase (AMPK)-ULK1 and autophagy pathways, whereas LAP controls G-CSF and GM-CSF expression to support MDSC development. Glycolytic signatures that include lactate dehydrogenase A correlate with high MDSCs and low T cells, and are associated with poor human TNBC outcome. Collectively, tumor glycolysis orchestrates a molecular network of the AMPK-ULK1, autophagy, and CEBPB pathways to affect MDSCs and maintain tumor immunosuppression.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Glucólisis , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Tolerancia Inmunológica , Células Supresoras de Origen Mieloide/inmunología , Neoplasias de la Mama Triple Negativas/inmunología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones
20.
J Clin Invest ; 128(2): 805-815, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29337305

RESUMEN

Programmed death-1 receptor (PD-L1, B7-H1) and programmed cell death protein 1 (PD-1) pathway blockade is a promising therapy for treating cancer. However, the mechanistic contribution of host and tumor PD-L1 and PD-1 signaling to the therapeutic efficacy of PD-L1 and PD-1 blockade remains elusive. Here, we evaluated 3 tumor-bearing mouse models that differ in their sensitivity to PD-L1 blockade and demonstrated a loss of therapeutic efficacy of PD-L1 blockade in immunodeficient mice and in PD-L1- and PD-1-deficient mice. In contrast, neither knockout nor overexpression of PD-L1 in tumor cells had an effect on PD-L1 blockade efficacy. Human and murine studies showed high levels of functional PD-L1 expression in dendritic cells and macrophages in the tumor microenvironments and draining lymph nodes. Additionally, expression of PD-L1 on dendritic cells and macrophages in ovarian cancer and melanoma patients correlated with the efficacy of treatment with either anti-PD-1 alone or in combination with anti-CTLA-4. Thus, PD-L1-expressing dendritic cells and macrophages may mechanistically shape and therapeutically predict clinical efficacy of PD-L1/PD-1 blockade.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Citocinas/metabolismo , Dendritas/metabolismo , Células Dendríticas/metabolismo , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/patología , Femenino , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Ganglios Linfáticos/patología , Macrófagos/metabolismo , Masculino , Melanoma/tratamiento farmacológico , Melanoma/patología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Neoplasias Ováricas/patología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Factores de Tiempo , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...