Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.140
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 36: 221-246, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29328786

RESUMEN

Researchers are intensifying efforts to understand the mechanisms by which changes in metabolic states influence differentiation programs. An emerging objective is to define how fluctuations in metabolites influence the epigenetic states that contribute to differentiation programs. This is because metabolites such as S-adenosylmethionine, acetyl-CoA, α-ketoglutarate, 2-hydroxyglutarate, and butyrate are donors, substrates, cofactors, and antagonists for the activities of epigenetic-modifying complexes and for epigenetic modifications. We discuss this topic from the perspective of specialized CD4+ T cells as well as effector and memory T cell differentiation programs. We also highlight findings from embryonic stem cells that give mechanistic insight into how nutrients processed through pathways such as glycolysis, glutaminolysis, and one-carbon metabolism regulate metabolite levels to influence epigenetic events and discuss similar mechanistic principles in T cells. Finally, we highlight how dysregulated environments, such as the tumor microenvironment, might alter programming events.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/inmunología , Metabolismo Energético , Epigénesis Genética , Animales , Biomarcadores , Regulación del Desarrollo de la Expresión Génica , Humanos , Neoplasias/etiología , Neoplasias/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
2.
Cell ; 185(1): 169-183.e19, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34963055

RESUMEN

Non-small cell lung cancers (NSCLCs) harboring KEAP1 mutations are often resistant to immunotherapy. Here, we show that KEAP1 targets EMSY for ubiquitin-mediated degradation to regulate homologous recombination repair (HRR) and anti-tumor immunity. Loss of KEAP1 in NSCLC induces stabilization of EMSY, producing a BRCAness phenotype, i.e., HRR defects and sensitivity to PARP inhibitors. Defective HRR contributes to a high tumor mutational burden that, in turn, is expected to prompt an innate immune response. Notably, EMSY accumulation suppresses the type I interferon response and impairs innate immune signaling, fostering cancer immune evasion. Activation of the type I interferon response in the tumor microenvironment using a STING agonist results in the engagement of innate and adaptive immune signaling and impairs the growth of KEAP1-mutant tumors. Our results suggest that targeting PARP and STING pathways, individually or in combination, represents a therapeutic strategy in NSCLC patients harboring alterations in KEAP1.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Interferón Tipo I/metabolismo , Neoplasias Pulmonares/inmunología , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Reparación del ADN por Recombinación/genética , Proteínas Represoras/metabolismo , Escape del Tumor/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Inmunidad Innata/genética , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Mutación , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Transducción de Señal/genética , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell ; 182(5): 1232-1251.e22, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32822576

RESUMEN

Lung cancer, the leading cause of cancer mortality, exhibits heterogeneity that enables adaptability, limits therapeutic success, and remains incompletely understood. Single-cell RNA sequencing (scRNA-seq) of metastatic lung cancer was performed using 49 clinical biopsies obtained from 30 patients before and during targeted therapy. Over 20,000 cancer and tumor microenvironment (TME) single-cell profiles exposed a rich and dynamic tumor ecosystem. scRNA-seq of cancer cells illuminated targetable oncogenes beyond those detected clinically. Cancer cells surviving therapy as residual disease (RD) expressed an alveolar-regenerative cell signature suggesting a therapy-induced primitive cell-state transition, whereas those present at on-therapy progressive disease (PD) upregulated kynurenine, plasminogen, and gap-junction pathways. Active T-lymphocytes and decreased macrophages were present at RD and immunosuppressive cell states characterized PD. Biological features revealed by scRNA-seq were biomarkers of clinical outcomes in independent cohorts. This study highlights how therapy-induced adaptation of the multi-cellular ecosystem of metastatic cancer shapes clinical outcomes.


Asunto(s)
Neoplasias Pulmonares/genética , Biomarcadores de Tumor/genética , Línea Celular , Ecosistema , Humanos , Neoplasias Pulmonares/patología , Macrófagos/patología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Linfocitos T/patología , Microambiente Tumoral/genética
4.
Cell ; 183(2): 377-394.e21, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32976798

RESUMEN

We employed scRNA sequencing to extensively characterize the cellular landscape of human liver from development to disease. Analysis of ∼212,000 cells representing human fetal, hepatocellular carcinoma (HCC), and mouse liver revealed remarkable fetal-like reprogramming of the tumor microenvironment. Specifically, the HCC ecosystem displayed features reminiscent of fetal development, including re-emergence of fetal-associated endothelial cells (PLVAP/VEGFR2) and fetal-like (FOLR2) tumor-associated macrophages. In a cross-species comparative analysis, we discovered remarkable similarity between mouse embryonic, fetal-liver, and tumor macrophages. Spatial transcriptomics further revealed a shared onco-fetal ecosystem between fetal liver and HCC. Furthermore, gene regulatory analysis, spatial transcriptomics, and in vitro functional assays implicated VEGF and NOTCH signaling in maintaining onco-fetal ecosystem. Taken together, we report a shared immunosuppressive onco-fetal ecosystem in fetal liver and HCC. Our results unravel a previously unexplored onco-fetal reprogramming of the tumor ecosystem, provide novel targets for therapeutic interventions in HCC, and open avenues for identifying similar paradigms in other cancers and disease.


Asunto(s)
Carcinoma Hepatocelular/patología , Células Endoteliales/metabolismo , Microambiente Tumoral/genética , Adulto , Animales , Carcinoma Hepatocelular/genética , Línea Celular , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Receptor 2 de Folato/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Hígado/patología , Neoplasias Hepáticas/genética , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Transcriptoma/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Cell ; 181(2): 442-459.e29, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32302573

RESUMEN

Single-cell RNA sequencing (scRNA-seq) is a powerful tool for defining cellular diversity in tumors, but its application toward dissecting mechanisms underlying immune-modulating therapies is scarce. We performed scRNA-seq analyses on immune and stromal populations from colorectal cancer patients, identifying specific macrophage and conventional dendritic cell (cDC) subsets as key mediators of cellular cross-talk in the tumor microenvironment. Defining comparable myeloid populations in mouse tumors enabled characterization of their response to myeloid-targeted immunotherapy. Treatment with anti-CSF1R preferentially depleted macrophages with an inflammatory signature but spared macrophage populations that in mouse and human expresses pro-angiogenic/tumorigenic genes. Treatment with a CD40 agonist antibody preferentially activated a cDC population and increased Bhlhe40+ Th1-like cells and CD8+ memory T cells. Our comprehensive analysis of key myeloid subsets in human and mouse identifies critical cellular interactions regulating tumor immunity and defines mechanisms underlying myeloid-targeted immunotherapies currently undergoing clinical testing.


Asunto(s)
Neoplasias del Colon/patología , Células Mieloides/metabolismo , Análisis de la Célula Individual/métodos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Secuencia de Bases/genética , Linfocitos T CD8-positivos/inmunología , China , Neoplasias del Colon/terapia , Neoplasias Colorrectales/patología , Células Dendríticas/inmunología , Femenino , Humanos , Inmunoterapia , Macrófagos/inmunología , Masculino , Ratones , Persona de Mediana Edad , Análisis de Secuencia de ARN/métodos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
6.
Nat Immunol ; 23(6): 971-984, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35624211

RESUMEN

Glioblastoma (GBM) is an incurable primary malignant brain cancer hallmarked with a substantial protumorigenic immune component. Knowledge of the GBM immune microenvironment during tumor evolution and standard of care treatments is limited. Using single-cell transcriptomics and flow cytometry, we unveiled large-scale comprehensive longitudinal changes in immune cell composition throughout tumor progression in an epidermal growth factor receptor-driven genetic mouse GBM model. We identified subsets of proinflammatory microglia in developing GBMs and anti-inflammatory macrophages and protumorigenic myeloid-derived suppressors cells in end-stage tumors, an evolution that parallels breakdown of the blood-brain barrier and extensive growth of epidermal growth factor receptor+ GBM cells. A similar relationship was found between microglia and macrophages in patient biopsies of low-grade glioma and GBM. Temozolomide decreased the accumulation of myeloid-derived suppressor cells, whereas concomitant temozolomide irradiation increased intratumoral GranzymeB+ CD8+T cells but also increased CD4+ regulatory T cells. These results provide a comprehensive and unbiased immune cellular landscape and its evolutionary changes during GBM progression.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Animales , Neoplasias Encefálicas/metabolismo , Receptores ErbB , Glioblastoma/metabolismo , Humanos , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Temozolomida/uso terapéutico , Microambiente Tumoral/genética
7.
Cell ; 179(2): 561-577.e22, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585088

RESUMEN

We performed the first proteogenomic characterization of hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) using paired tumor and adjacent liver tissues from 159 patients. Integrated proteogenomic analyses revealed consistency and discordance among multi-omics, activation status of key signaling pathways, and liver-specific metabolic reprogramming in HBV-related HCC. Proteomic profiling identified three subgroups associated with clinical and molecular attributes including patient survival, tumor thrombus, genetic profile, and the liver-specific proteome. These proteomic subgroups have distinct features in metabolic reprogramming, microenvironment dysregulation, cell proliferation, and potential therapeutics. Two prognostic biomarkers, PYCR2 and ADH1A, related to proteomic subgrouping and involved in HCC metabolic reprogramming, were identified. CTNNB1 and TP53 mutation-associated signaling and metabolic profiles were revealed, among which mutated CTNNB1-associated ALDOA phosphorylation was validated to promote glycolysis and cell proliferation. Our study provides a valuable resource that significantly expands the knowledge of HBV-related HCC and may eventually benefit clinical practice.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virología , Fructosa-Bifosfato Aldolasa/genética , Virus de la Hepatitis B , Hepatitis B Crónica/complicaciones , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virología , Proteogenómica/métodos , beta Catenina/genética , Animales , Proliferación Celular , Estudios de Cohortes , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Microambiente Tumoral/genética
8.
Cell ; 179(4): 964-983.e31, 2019 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31675502

RESUMEN

To elucidate the deregulated functional modules that drive clear cell renal cell carcinoma (ccRCC), we performed comprehensive genomic, epigenomic, transcriptomic, proteomic, and phosphoproteomic characterization of treatment-naive ccRCC and paired normal adjacent tissue samples. Genomic analyses identified a distinct molecular subgroup associated with genomic instability. Integration of proteogenomic measurements uniquely identified protein dysregulation of cellular mechanisms impacted by genomic alterations, including oxidative phosphorylation-related metabolism, protein translation processes, and phospho-signaling modules. To assess the degree of immune infiltration in individual tumors, we identified microenvironment cell signatures that delineated four immune-based ccRCC subtypes characterized by distinct cellular pathways. This study reports a large-scale proteogenomic analysis of ccRCC to discern the functional impact of genomic alterations and provides evidence for rational treatment selection stemming from ccRCC pathobiology.


Asunto(s)
Carcinoma de Células Renales/genética , Proteínas de Neoplasias/genética , Proteogenómica , Transcriptoma/genética , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Supervivencia sin Enfermedad , Exoma/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Genoma Humano/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/inmunología , Fosforilación Oxidativa , Fosforilación/genética , Transducción de Señal/genética , Transcriptoma/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Secuenciación del Exoma
9.
Cell ; 178(4): 835-849.e21, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31327527

RESUMEN

Diverse genetic, epigenetic, and developmental programs drive glioblastoma, an incurable and poorly understood tumor, but their precise characterization remains challenging. Here, we use an integrative approach spanning single-cell RNA-sequencing of 28 tumors, bulk genetic and expression analysis of 401 specimens from the The Cancer Genome Atlas (TCGA), functional approaches, and single-cell lineage tracing to derive a unified model of cellular states and genetic diversity in glioblastoma. We find that malignant cells in glioblastoma exist in four main cellular states that recapitulate distinct neural cell types, are influenced by the tumor microenvironment, and exhibit plasticity. The relative frequency of cells in each state varies between glioblastoma samples and is influenced by copy number amplifications of the CDK4, EGFR, and PDGFRA loci and by mutations in the NF1 locus, which each favor a defined state. Our work provides a blueprint for glioblastoma, integrating the malignant cell programs, their plasticity, and their modulation by genetic drivers.


Asunto(s)
Neoplasias Encefálicas/genética , Plasticidad de la Célula/genética , Glioblastoma/genética , Adolescente , Anciano , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Linaje de la Célula/genética , Niño , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Heterogeneidad Genética , Glioblastoma/patología , Xenoinjertos , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Persona de Mediana Edad , Mutación , RNA-Seq , Análisis de la Célula Individual/métodos , Microambiente Tumoral/genética
10.
Cell ; 179(4): 829-845.e20, 2019 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-31675496

RESUMEN

The immune microenvironment of hepatocellular carcinoma (HCC) is poorly characterized. Combining two single-cell RNA sequencing technologies, we produced transcriptomes of CD45+ immune cells for HCC patients from five immune-relevant sites: tumor, adjacent liver, hepatic lymph node (LN), blood, and ascites. A cluster of LAMP3+ dendritic cells (DCs) appeared to be the mature form of conventional DCs and possessed the potential to migrate from tumors to LNs. LAMP3+ DCs also expressed diverse immune-relevant ligands and exhibited potential to regulate multiple subtypes of lymphocytes. Of the macrophages in tumors that exhibited distinct transcriptional states, tumor-associated macrophages (TAMs) were associated with poor prognosis, and we established the inflammatory role of SLC40A1 and GPNMB in these cells. Further, myeloid and lymphoid cells in ascites were predominantly linked to tumor and blood origins, respectively. The dynamic properties of diverse CD45+ cell types revealed by this study add new dimensions to the immune landscape of HCC.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Proteínas de Transporte de Catión/genética , Inflamación/inmunología , Neoplasias Hepáticas/inmunología , Glicoproteínas de Membrana/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Comunicación Celular/genética , Comunicación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Células Dendríticas/inmunología , Células Dendríticas/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/patología , Antígenos Comunes de Leucocito/inmunología , Hígado/inmunología , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Linfocitos/inmunología , Linfocitos/patología , Proteínas de Membrana de los Lisosomas/genética , Macrófagos/inmunología , Macrófagos/patología , Células Mieloides/inmunología , Células Mieloides/patología , Proteínas de Neoplasias/genética , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcriptoma/genética , Transcriptoma/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
11.
Nat Immunol ; 22(6): 769-780, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34017122

RESUMEN

Progression and persistence of malignancies are influenced by the local tumor microenvironment, and future eradication of currently incurable tumors will, in part, hinge on our understanding of malignant cell biology in the context of their nourishing surroundings. Here, we generated paired single-cell transcriptomic datasets of tumor cells and the bone marrow immune and stromal microenvironment in multiple myeloma. These analyses identified myeloma-specific inflammatory mesenchymal stromal cells, which spatially colocalized with tumor cells and immune cells and transcribed genes involved in tumor survival and immune modulation. Inflammatory stromal cell signatures were driven by stimulation with proinflammatory cytokines, and analyses of immune cell subsets suggested interferon-responsive effector T cell and CD8+ stem cell memory T cell populations as potential sources of stromal cell-activating cytokines. Tracking stromal inflammation in individuals over time revealed that successful antitumor induction therapy is unable to revert bone marrow inflammation, predicting a role for mesenchymal stromal cells in disease persistence.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Células Madre Mesenquimatosas/inmunología , Mieloma Múltiple/inmunología , Recurrencia Local de Neoplasia/inmunología , Microambiente Tumoral/inmunología , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Médula Ósea/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Masculino , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/prevención & control , Cultivo Primario de Células , Estudios Prospectivos , RNA-Seq , Análisis de la Célula Individual , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
12.
Cell ; 173(2): 305-320.e10, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29625049

RESUMEN

The Cancer Genome Atlas (TCGA) has catalyzed systematic characterization of diverse genomic alterations underlying human cancers. At this historic junction marking the completion of genomic characterization of over 11,000 tumors from 33 cancer types, we present our current understanding of the molecular processes governing oncogenesis. We illustrate our insights into cancer through synthesis of the findings of the TCGA PanCancer Atlas project on three facets of oncogenesis: (1) somatic driver mutations, germline pathogenic variants, and their interactions in the tumor; (2) the influence of the tumor genome and epigenome on transcriptome and proteome; and (3) the relationship between tumor and the microenvironment, including implications for drugs targeting driver events and immunotherapies. These results will anchor future characterization of rare and common tumor types, primary and relapsed tumors, and cancers across ancestry groups and will guide the deployment of clinical genomic sequencing.


Asunto(s)
Carcinogénesis/genética , Genómica , Neoplasias/patología , Reparación del ADN/genética , Bases de Datos Genéticas , Genes Relacionados con las Neoplasias , Humanos , Redes y Vías Metabólicas/genética , Inestabilidad de Microsatélites , Mutación , Neoplasias/genética , Neoplasias/inmunología , Transcriptoma , Microambiente Tumoral/genética
13.
Nat Immunol ; 21(3): 287-297, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31932812

RESUMEN

Cancer cells subvert immune surveillance through inhibition of T cell effector function. Elucidation of the mechanism of T cell dysfunction is therefore central to cancer immunotherapy. Here, we report that dual specificity phosphatase 2 (DUSP2; also known as phosphatase of activated cells 1, PAC1) acts as an immune checkpoint in T cell antitumor immunity. PAC1 is selectively upregulated in exhausted tumor-infiltrating lymphocytes and is associated with poor prognosis of patients with cancer. PAC1hi effector T cells lose their proliferative and effector capacities and convert into exhausted T cells. Deletion of PAC1 enhances immune responses and reduces cancer susceptibility in mice. Through activation of EGR1, excessive reactive oxygen species in the tumor microenvironment induce expression of PAC1, which recruits the Mi-2ß nucleosome-remodeling and histone-deacetylase complex, eventually leading to chromatin remodeling of effector T cells. Our study demonstrates that PAC1 is an epigenetic immune regulator and highlights the importance of targeting PAC1 in cancer immunotherapy.


Asunto(s)
Fosfatasa 2 de Especificidad Dual/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , Animales , Cromatina/genética , Cromatina/metabolismo , Fosfatasa 2 de Especificidad Dual/deficiencia , Fosfatasa 2 de Especificidad Dual/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Humanos , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/genética , Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Regulación hacia Arriba
14.
Immunity ; 56(10): 2188-2205, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37820582

RESUMEN

The cancer-immunity cycle provides a framework to understand the series of events that generate anti-cancer immune responses. It emphasizes the iterative nature of the response where the killing of tumor cells by T cells initiates subsequent rounds of antigen presentation and T cell stimulation, maintaining active immunity and adapting it to tumor evolution. Any step of the cycle can become rate-limiting, rendering the immune system unable to control tumor growth. Here, we update the cancer-immunity cycle based on the remarkable progress of the past decade. Understanding the mechanism of checkpoint inhibition has evolved, as has our view of dendritic cells in sustaining anti-tumor immunity. We additionally account for the role of the tumor microenvironment in facilitating, not just suppressing, the anti-cancer response, and discuss the importance of considering a tumor's immunological phenotype, the "immunotype". While these new insights add some complexity to the cycle, they also provide new targets for research and therapeutic intervention.


Asunto(s)
Inmunoterapia , Neoplasias , Humanos , Neoplasias/genética , Neoplasias/terapia , Neoplasias/metabolismo , Linfocitos T , Presentación de Antígeno , Genotipo , Microambiente Tumoral/genética
15.
Annu Rev Cell Dev Biol ; 34: 333-355, 2018 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-30028641

RESUMEN

Stellate cells are resident lipid-storing cells of the pancreas and liver that transdifferentiate to a myofibroblastic state in the context of tissue injury. Beyond having roles in tissue homeostasis, stellate cells are increasingly implicated in pathological fibrogenic and inflammatory programs that contribute to tissue fibrosis and that constitute a growth-permissive tumor microenvironment. Although the capacity of stellate cells for extracellular matrix production and remodeling has long been appreciated, recent research efforts have demonstrated diverse roles for stellate cells in regulation of epithelial cell fate, immune modulation, and tissue health. Our present understanding of stellate cell biology in health and disease is discussed here, as are emerging means to target these multifaceted cells for therapeutic benefit.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Inflamación/genética , Neoplasias/genética , Células Estrelladas Pancreáticas/metabolismo , Transdiferenciación Celular/genética , Células Estrelladas Hepáticas/patología , Humanos , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Neoplasias/patología , Páncreas/lesiones , Páncreas/metabolismo , Páncreas/patología , Células Estrelladas Pancreáticas/patología , Microambiente Tumoral/genética , Cicatrización de Heridas
16.
Nat Immunol ; 20(11): 1494-1505, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31611701

RESUMEN

Immunotherapy has transformed cancer treatment. However, current immunotherapy modalities face various limitations. In the present study, we developed multiplexed activation of endogenous genes as an immunotherapy (MAEGI), a new form of immunotherapy that elicits antitumor immunity through multiplexed activation of endogenous genes in tumors. We leveraged CRISPR activation (CRISPRa) to directly augment the in situ expression of endogenous genes, and thereby the presentation of tumor antigens, leading to dramatic antitumor immune responses. Deploying this as a cell-based vaccination strategy showed efficacy in both prophylactic and therapeutic settings. Intratumoral adeno-associated virus delivery of CRISPRa libraries elicited strong antitumor immunity across multiple cancer types. Precision targeting of mutated gene sets eradicated a large fraction of established tumors at both local and distant sites. This treatment modality led to alterations in the tumor microenvironment, marked by enhanced T cell infiltration and antitumor immune signatures. Multiplexed endogenous gene activation is a versatile and highly scalable strategy to elicit potent immune responses against cancer, distinct from all existing cancer therapies.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Terapia Genética/métodos , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Animales , Presentación de Antígeno/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Terapia Combinada/métodos , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células HEK293 , Humanos , Inyecciones Intralesiones , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Neoplasias/genética , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
17.
Nat Immunol ; 20(7): 835-851, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31160797

RESUMEN

How tumor cells genetically lose antigenicity and evade immune checkpoints remains largely elusive. We report that tissue-specific expression of the human long noncoding RNA LINK-A in mouse mammary glands initiates metastatic mammary gland tumors, which phenotypically resemble human triple-negative breast cancer (TNBC). LINK-A expression facilitated crosstalk between phosphatidylinositol-(3,4,5)-trisphosphate and inhibitory G-protein-coupled receptor (GPCR) pathways, attenuating protein kinase A-mediated phosphorylation of the E3 ubiquitin ligase TRIM71. Consequently, LINK-A expression enhanced K48-polyubiquitination-mediated degradation of the antigen peptide-loading complex (PLC) and intrinsic tumor suppressors Rb and p53. Treatment with LINK-A locked nucleic acids or GPCR antagonists stabilized the PLC components, Rb and p53, and sensitized mammary gland tumors to immune checkpoint blockers. Patients with programmed ccll death protein-1(PD-1) blockade-resistant TNBC exhibited elevated LINK-A levels and downregulated PLC components. Hence we demonstrate lncRNA-dependent downregulation of antigenicity and intrinsic tumor suppression, which provides the basis for developing combinational immunotherapy treatment regimens and early TNBC prevention.


Asunto(s)
Presentación de Antígeno/inmunología , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Neoplasias/inmunología , Oncogenes , ARN Largo no Codificante/genética , Escape del Tumor/genética , Escape del Tumor/inmunología , Adenoma/genética , Adenoma/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nat Rev Genet ; 24(5): 295-313, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36494509

RESUMEN

The natural history of cancers can be understood through the lens of evolution given that the driving forces of cancer development are mutation and selection of fitter clones. Cancer growth and progression are spatial processes that involve the breakdown of normal tissue organization, invasion and metastasis. For these reasons, spatial patterns are an integral part of histological tumour grading and staging as they measure the progression from normal to malignant disease. Furthermore, tumour cells are part of an ecosystem of tumour cells and their surrounding tumour microenvironment. A range of new spatial genomic, transcriptomic and proteomic technologies offers new avenues for the study of cancer evolution with great molecular and spatial detail. These methods enable precise characterizations of the tumour microenvironment, cellular interactions therein and micro-anatomical structures. In conjunction with spatial genomics, it emerges that tumours and microenvironments co-evolve, which helps explain observable patterns of heterogeneity and offers new routes for therapeutic interventions.


Asunto(s)
Neoplasias , Proteómica , Humanos , Ecosistema , Neoplasias/genética , Neoplasias/patología , Genómica , Microambiente Tumoral/genética
19.
Mol Cell ; 81(5): 940-952.e5, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33434504

RESUMEN

STING-dependent cytosolic DNA sensing in dendritic cells (DCs) initiates antitumor immune responses, but how STING signaling is metabolically regulated in the tumor microenvironment remains unknown. Here, we show that oxidative stress is required for STING-induced DC antitumor function through a process that directs SUMO-specific protease 3 (SENP3) activity. DC-specific deletion of Senp3 drives tumor progression by blunting STING-dependent type-I interferon (IFN) signaling in DCs and dampening antitumor immune responses. DC-derived reactive oxygen species (ROS) trigger SENP3 accumulation and the SENP3-IFI204 interaction, thereby catalyzing IFI204 deSUMOylation and boosting STING signaling activation in mice. Consistently, SENP3 senses ROS to facilitate STING-dependent DC activity in tissue samples from colorectal cancer patients. Our results reveal that oxidative stress as a metabolic regulator promotes STING-mediated DC antitumor immune responses and highlights SENP3 as an overflow valve for STING signaling induction in the metabolically abnormal tumor microenvironment.


Asunto(s)
Neoplasias Colorrectales/genética , Cisteína Endopeptidasas/genética , Células Dendríticas/inmunología , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Aloinjertos , Animales , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Cisteína Endopeptidasas/inmunología , Células Dendríticas/patología , Femenino , Células HEK293 , Humanos , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/inmunología , Estrés Oxidativo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Análisis de Supervivencia , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
20.
Genes Dev ; 35(13-14): 963-975, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34168038

RESUMEN

Autophagy inhibitors are currently being evaluated in clinical trials for the treatment of diverse cancers, largely due to their ability to impede tumor cell survival and metabolic adaptation. More recently, there is growing interest in whether and how modulating autophagy in the host stroma influences tumorigenesis. Fibroblasts play prominent roles in cancer initiation and progression, including depositing type 1 collagen and other extracellular matrix (ECM) components, thereby stiffening the surrounding tissue to enhance tumor cell proliferation and survival, as well as secreting cytokines that modulate angiogenesis and the immune microenvironment. This constellation of phenotypes, pathologically termed desmoplasia, heralds poor prognosis and reduces patient survival. Using mouse mammary cancer models and syngeneic transplantation assays, we demonstrate that genetic ablation of stromal fibroblast autophagy significantly impedes fundamental elements of the stromal desmoplastic response, including collagen and proinflammatory cytokine secretion, extracellular matrix stiffening, and neoangiogenesis. As a result, autophagy in stromal fibroblasts is required for mammary tumor growth in vivo, even when the cancer cells themselves remain autophagy-competent . We propose the efficacy of autophagy inhibition is shaped by this ability of host stromal fibroblast autophagy to support tumor desmoplasia.


Asunto(s)
Células del Estroma , Microambiente Tumoral , Animales , Autofagia/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Fibroblastos/metabolismo , Humanos , Ratones , Microambiente Tumoral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA