Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113.188
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(9): 2126-2128, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38670070

RESUMEN

The landscape of the intratumoral microbiome in tumor metastases is largely unchartered. In this issue of Cell, Voest et al. profiled the tumor metastasis-associated microbiome in a pancancer cohort of 4,160 biopsies from 26 cancer types. This dataset offers a useful resource for understanding the role of the microbiome in metastatic cancers.


Asunto(s)
Microbiota , Metástasis de la Neoplasia , Humanos , Neoplasias/patología , Neoplasias/microbiología
2.
Cell ; 187(9): 2324-2335.e19, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38599211

RESUMEN

Microbial communities are resident to multiple niches of the human body and are important modulators of the host immune system and responses to anticancer therapies. Recent studies have shown that complex microbial communities are present within primary tumors. To investigate the presence and relevance of the microbiome in metastases, we integrated mapping and assembly-based metagenomics, genomics, transcriptomics, and clinical data of 4,160 metastatic tumor biopsies. We identified organ-specific tropisms of microbes, enrichments of anaerobic bacteria in hypoxic tumors, associations between microbial diversity and tumor-infiltrating neutrophils, and the association of Fusobacterium with resistance to immune checkpoint blockade (ICB) in lung cancer. Furthermore, longitudinal tumor sampling revealed temporal evolution of the microbial communities and identified bacteria depleted upon ICB. Together, we generated a pan-cancer resource of the metastatic tumor microbiome that may contribute to advancing treatment strategies.


Asunto(s)
Microbiota , Metástasis de la Neoplasia , Neoplasias , Humanos , Neoplasias/microbiología , Neoplasias/patología , Metagenómica/métodos , Neoplasias Pulmonares/microbiología , Neoplasias Pulmonares/patología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Neutrófilos/inmunología , Microambiente Tumoral , Bacterias/genética , Bacterias/clasificación
3.
Cell ; 186(8): 1564-1579, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37059065

RESUMEN

Most cancer-associated deaths occur due to metastasis, yet our understanding of metastasis as an evolving, heterogeneous, systemic disease and of how to effectively treat it is still emerging. Metastasis requires the acquisition of a succession of traits to disseminate, variably enter and exit dormancy, and colonize distant organs. The success of these events is driven by clonal selection, the potential of metastatic cells to dynamically transition into distinct states, and their ability to co-opt the immune environment. Here, we review the main principles of metastasis and highlight emerging opportunities to develop more effective therapies for metastatic cancer.


Asunto(s)
Neoplasias , Humanos , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/terapia , Neoplasias/patología , Neoplasias/terapia
4.
Cell ; 186(26): 5719-5738.e28, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38056463

RESUMEN

Tumor-associated hydrocephalus (TAH) is a common and lethal complication of brain metastases. Although other factors beyond mechanical obstructions have been suggested, the exact mechanisms are unknown. Using single-nucleus RNA sequencing and spatial transcriptomics, we find that a distinct population of mast cells locate in the choroid plexus and dramatically increase during TAH. Genetic fate tracing and intracranial mast-cell-specific tryptase knockout showed that choroid plexus mast cells (CPMCs) disrupt cilia of choroid plexus epithelia via the tryptase-PAR2-FoxJ1 pathway and consequently increase cerebrospinal fluid production. Mast cells are also found in the human choroid plexus. Levels of tryptase in cerebrospinal fluid are closely associated with clinical severity of TAH. BMS-262084, an inhibitor of tryptase, can cross the blood-brain barrier, inhibit TAH in vivo, and alleviate mast-cell-induced damage of epithelial cilia in a human pluripotent stem-cell-derived choroid plexus organoid model. Collectively, we uncover the function of CPMCs and provide an attractive therapy for TAH.


Asunto(s)
Neoplasias Encefálicas , Plexo Coroideo , Hidrocefalia , Mastocitos , Humanos , Neoplasias Encefálicas/secundario , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Hidrocefalia/metabolismo , Hidrocefalia/patología , Mastocitos/metabolismo , Mastocitos/patología , Triptasas/líquido cefalorraquídeo , Metástasis de la Neoplasia/patología
5.
Cell ; 185(3): 563-575.e11, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35120664

RESUMEN

Metastatic progression is the main cause of death in cancer patients, whereas the underlying genomic mechanisms driving metastasis remain largely unknown. Here, we assembled MSK-MET, a pan-cancer cohort of over 25,000 patients with metastatic diseases. By analyzing genomic and clinical data from this cohort, we identified associations between genomic alterations and patterns of metastatic dissemination across 50 tumor types. We found that chromosomal instability is strongly correlated with metastatic burden in some tumor types, including prostate adenocarcinoma, lung adenocarcinoma, and HR+/HER2+ breast ductal carcinoma, but not in others, including colorectal cancer and high-grade serous ovarian cancer, where copy-number alteration patterns may be established early in tumor development. We also identified somatic alterations associated with metastatic burden and specific target organs. Our data offer a valuable resource for the investigation of the biological basis for metastatic spread and highlight the complex role of chromosomal instability in cancer progression.


Asunto(s)
Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Estudios de Cohortes , Femenino , Humanos , Masculino , Especificidad de Órganos/genética , Estudios Prospectivos
6.
Cell ; 185(8): 1292-1294, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35427497

RESUMEN

Tumors contain bacteria, but the functional significance of this tumor microbiota is not appreciated. Fu et al. show that bacteria within breast tumor cells contribute to metastasis, in part, by enhancing tumor cell survival to mechanical fluid shear stress as would be found in the circulation.


Asunto(s)
Bacterias , Neoplasias de la Mama , Metástasis de la Neoplasia , Neoplasias de la Mama/microbiología , Neoplasias de la Mama/patología , Supervivencia Celular , Femenino , Humanos , Estrés Mecánico
7.
Cell ; 185(8): 1356-1372.e26, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35395179

RESUMEN

Tumor-resident intracellular microbiota is an emerging tumor component that has been documented for a variety of cancer types with unclear biological functions. Here, we explored the functional significance of these intratumor bacteria, primarily using a murine spontaneous breast-tumor model MMTV-PyMT. We found that depletion of intratumor bacteria significantly reduced lung metastasis without affecting primary tumor growth. During metastatic colonization, intratumor bacteria carried by circulating tumor cells promoted host-cell survival by enhancing resistance to fluid shear stress by reorganizing actin cytoskeleton. We further showed that intratumor administration of selected bacteria strains isolated from tumor-resident microbiota promoted metastasis in two murine tumor models with significantly different levels of metastasis potential. Our findings suggest that tumor-resident microbiota, albeit at low biomass, play an important role in promoting cancer metastasis, intervention of which might therefore be worth exploring for advancing oncology care.


Asunto(s)
Neoplasias de la Mama , Microbiota , Metástasis de la Neoplasia , Animales , Neoplasias de la Mama/microbiología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/patología , Ratones , Células Neoplásicas Circulantes/patología
8.
Nat Immunol ; 25(5): 916-924, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38698238

RESUMEN

B cells and T cells are important components of the adaptive immune system and mediate anticancer immunity. The T cell landscape in cancer is well characterized, but the contribution of B cells to anticancer immunosurveillance is less well explored. Here we show an integrative analysis of the B cell and T cell receptor repertoire from individuals with metastatic breast cancer and individuals with early breast cancer during neoadjuvant therapy. Using immune receptor, RNA and whole-exome sequencing, we show that both B cell and T cell responses seem to coevolve with the metastatic cancer genomes and mirror tumor mutational and neoantigen architecture. B cell clones associated with metastatic immunosurveillance and temporal persistence were more expanded and distinct from site-specific clones. B cell clonal immunosurveillance and temporal persistence are predictable from the clonal structure, with higher-centrality B cell antigen receptors more likely to be detected across multiple metastases or across time. This predictability was generalizable across other immune-mediated disorders. This work lays a foundation for prioritizing antibody sequences for therapeutic targeting in cancer.


Asunto(s)
Linfocitos B , Neoplasias de la Mama , Vigilancia Inmunológica , Humanos , Femenino , Neoplasias de la Mama/inmunología , Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Linfocitos T/inmunología , Monitorización Inmunológica , Secuenciación del Exoma , Antígenos de Neoplasias/inmunología , Metástasis de la Neoplasia , Células Clonales
9.
Cell ; 184(6): 1650-1650.e1, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33740457

RESUMEN

Understanding how tumors grow and evolve over time is crucial to help shed light on the underlying reasons why treatments fail and tumors metastasize. This SnapShot provides a brief introduction into the main concepts of tumor evolution. To view this SnapShot, open or download the PDF.


Asunto(s)
Neoplasias/patología , Humanos , Mutación/genética , Metástasis de la Neoplasia , Neoplasias/genética
10.
Cell ; 184(8): 2033-2052.e21, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33765443

RESUMEN

Metastasis is the leading cause of cancer-related deaths, and greater knowledge of the metastatic microenvironment is necessary to effectively target this process. Microenvironmental changes occur at distant sites prior to clinically detectable metastatic disease; however, the key niche regulatory signals during metastatic progression remain poorly characterized. Here, we identify a core immune suppression gene signature in pre-metastatic niche formation that is expressed predominantly by myeloid cells. We target this immune suppression program by utilizing genetically engineered myeloid cells (GEMys) to deliver IL-12 to modulate the metastatic microenvironment. Our data demonstrate that IL12-GEMy treatment reverses immune suppression in the pre-metastatic niche by activating antigen presentation and T cell activation, resulting in reduced metastatic and primary tumor burden and improved survival of tumor-bearing mice. We demonstrate that IL12-GEMys can functionally modulate the core program of immune suppression in the pre-metastatic niche to successfully rebalance the dysregulated metastatic microenvironment in cancer.


Asunto(s)
Terapia de Inmunosupresión , Células Mieloides/metabolismo , Inmunidad Adaptativa , Animales , Línea Celular Tumoral , Ingeniería Genética , Humanos , Interleucina-12/genética , Interleucina-12/metabolismo , Pulmón/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/citología , Células Mieloides/inmunología , Metástasis de la Neoplasia , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral
11.
Cell ; 184(9): 2471-2486.e20, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33878291

RESUMEN

Metastasis has been considered as the terminal step of tumor progression. However, recent genomic studies suggest that many metastases are initiated by further spread of other metastases. Nevertheless, the corresponding pre-clinical models are lacking, and underlying mechanisms are elusive. Using several approaches, including parabiosis and an evolving barcode system, we demonstrated that the bone microenvironment facilitates breast and prostate cancer cells to further metastasize and establish multi-organ secondary metastases. We uncovered that this metastasis-promoting effect is driven by epigenetic reprogramming that confers stem cell-like properties on cancer cells disseminated from bone lesions. Furthermore, we discovered that enhanced EZH2 activity mediates the increased stemness and metastasis capacity. The same findings also apply to single cell-derived populations, indicating mechanisms distinct from clonal selection. Taken together, our work revealed an unappreciated role of the bone microenvironment in metastasis evolution and elucidated an epigenomic reprogramming process driving terminal-stage, multi-organ metastases.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , Microambiente Tumoral , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cell ; 183(2): 395-410.e19, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33007268

RESUMEN

Collective metastasis is defined as the cohesive migration and metastasis of multicellular tumor cell clusters. Disrupting various cell adhesion genes markedly reduces cluster formation and colonization efficiency, yet the downstream signals transmitted by clustering remain largely unknown. Here, we use mouse and human breast cancer models to identify a collective signal generated by tumor cell clusters supporting metastatic colonization. We show that tumor cell clusters produce the growth factor epigen and concentrate it within nanolumina-intercellular compartments sealed by cell-cell junctions and lined with microvilli-like protrusions. Epigen knockdown profoundly reduces metastatic outgrowth and switches clusters from a proliferative to a collective migratory state. Tumor cell clusters from basal-like 2, but not mesenchymal-like, triple-negative breast cancer cell lines have increased epigen expression, sealed nanolumina, and impaired outgrowth upon nanolumenal junction disruption. We propose that nanolumenal signaling could offer a therapeutic target for aggressive metastatic breast cancers.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Uniones Intercelulares/patología , Metástasis de la Neoplasia/fisiopatología , Animales , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Epigen/metabolismo , Transición Epitelial-Mesenquimal/genética , Humanos , Ratones , Células Neoplásicas Circulantes/patología , Transducción de Señal/fisiología , Neoplasias de la Mama Triple Negativas/patología
13.
Cell ; 181(7): 1626-1642.e20, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32470397

RESUMEN

Brain malignancies can either originate from within the CNS (gliomas) or invade from other locations in the body (metastases). A highly immunosuppressive tumor microenvironment (TME) influences brain tumor outgrowth. Whether the TME is predominantly shaped by the CNS micromilieu or by the malignancy itself is unknown, as is the diversity, origin, and function of CNS tumor-associated macrophages (TAMs). Here, we have mapped the leukocyte landscape of brain tumors using high-dimensional single-cell profiling (CyTOF). The heterogeneous composition of tissue-resident and invading immune cells within the TME alone permitted a clear distinction between gliomas and brain metastases (BrM). The glioma TME presented predominantly with tissue-resident, reactive microglia, whereas tissue-invading leukocytes accumulated in BrM. Tissue-invading TAMs showed a distinctive signature trajectory, revealing tumor-driven instruction along with contrasting lymphocyte activation and exhaustion. Defining the specific immunological signature of brain tumors can facilitate the rational design of targeted immunotherapy strategies.


Asunto(s)
Neoplasias Encefálicas/inmunología , Leucocitos/inmunología , Microambiente Tumoral/inmunología , Neoplasias Encefálicas/patología , Femenino , Glioma/patología , Humanos , Inmunoterapia , Leucocitos/metabolismo , Leucocitos/fisiología , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Microglía/patología , Metástasis de la Neoplasia/patología
14.
Cell ; 176(1-2): 98-112.e14, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30633912

RESUMEN

The ability of circulating tumor cells (CTCs) to form clusters has been linked to increased metastatic potential. Yet biological features and vulnerabilities of CTC clusters remain largely unknown. Here, we profile the DNA methylation landscape of single CTCs and CTC clusters from breast cancer patients and mouse models on a genome-wide scale. We find that binding sites for stemness- and proliferation-associated transcription factors are specifically hypomethylated in CTC clusters, including binding sites for OCT4, NANOG, SOX2, and SIN3A, paralleling embryonic stem cell biology. Among 2,486 FDA-approved compounds, we identify Na+/K+ ATPase inhibitors that enable the dissociation of CTC clusters into single cells, leading to DNA methylation remodeling at critical sites and metastasis suppression. Thus, our results link CTC clustering to specific changes in DNA methylation that promote stemness and metastasis and point to cluster-targeting compounds to suppress the spread of cancer.


Asunto(s)
Neoplasias de la Mama/genética , Metástasis de la Neoplasia/genética , Células Neoplásicas Circulantes/patología , Animales , Neoplasias de la Mama/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN/fisiología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Proteína Homeótica Nanog/metabolismo , Metástasis de la Neoplasia/fisiopatología , Células Neoplásicas Circulantes/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción SOXB1/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3
15.
Cell ; 179(7): 1661-1676.e19, 2019 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-31835038

RESUMEN

Reliable detection of disseminated tumor cells and of the biodistribution of tumor-targeting therapeutic antibodies within the entire body has long been needed to better understand and treat cancer metastasis. Here, we developed an integrated pipeline for automated quantification of cancer metastases and therapeutic antibody targeting, named DeepMACT. First, we enhanced the fluorescent signal of cancer cells more than 100-fold by applying the vDISCO method to image metastasis in transparent mice. Second, we developed deep learning algorithms for automated quantification of metastases with an accuracy matching human expert manual annotation. Deep learning-based quantification in 5 different metastatic cancer models including breast, lung, and pancreatic cancer with distinct organotropisms allowed us to systematically analyze features such as size, shape, spatial distribution, and the degree to which metastases are targeted by a therapeutic monoclonal antibody in entire mice. DeepMACT can thus considerably improve the discovery of effective antibody-based therapeutics at the pre-clinical stage. VIDEO ABSTRACT.


Asunto(s)
Anticuerpos/uso terapéutico , Aprendizaje Profundo , Diagnóstico por Computador/métodos , Quimioterapia Asistida por Computador/métodos , Neoplasias/patología , Animales , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Programas Informáticos , Microambiente Tumoral
16.
Cell ; 178(2): 330-345.e22, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31257027

RESUMEN

For tumors to progress efficiently, cancer cells must overcome barriers of oxidative stress. Although dietary antioxidant supplementation or activation of endogenous antioxidants by NRF2 reduces oxidative stress and promotes early lung tumor progression, little is known about its effect on lung cancer metastasis. Here, we show that long-term supplementation with the antioxidants N-acetylcysteine and vitamin E promotes KRAS-driven lung cancer metastasis. The antioxidants stimulate metastasis by reducing levels of free heme and stabilizing the transcription factor BACH1. BACH1 activates transcription of Hexokinase 2 and Gapdh and increases glucose uptake, glycolysis rates, and lactate secretion, thereby stimulating glycolysis-dependent metastasis of mouse and human lung cancer cells. Targeting BACH1 normalized glycolysis and prevented antioxidant-induced metastasis, while increasing endogenous BACH1 expression stimulated glycolysis and promoted metastasis, also in the absence of antioxidants. We conclude that BACH1 stimulates glycolysis-dependent lung cancer metastasis and that BACH1 is activated under conditions of reduced oxidative stress.


Asunto(s)
Antioxidantes/farmacología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Glucólisis/efectos de los fármacos , Neoplasias Pulmonares/patología , Animales , Antioxidantes/administración & dosificación , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Movimiento Celular/efectos de los fármacos , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Hemo/metabolismo , Hexoquinasa/antagonistas & inhibidores , Hexoquinasa/genética , Hexoquinasa/metabolismo , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/mortalidad , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Factor 2 Relacionado con NF-E2/metabolismo , Metástasis de la Neoplasia , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo
17.
Cell ; 178(2): 316-329.e18, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31257023

RESUMEN

Approximately 30% of human lung cancers acquire mutations in either Keap1 or Nfe2l2, resulting in the stabilization of Nrf2, the Nfe2l2 gene product, which controls oxidative homeostasis. Here, we show that heme triggers the degradation of Bach1, a pro-metastatic transcription factor, by promoting its interaction with the ubiquitin ligase Fbxo22. Nrf2 accumulation in lung cancers causes the stabilization of Bach1 by inducing Ho1, the enzyme catabolizing heme. In mouse models of lung cancers, loss of Keap1 or Fbxo22 induces metastasis in a Bach1-dependent manner. Pharmacological inhibition of Ho1 suppresses metastasis in a Fbxo22-dependent manner. Human metastatic lung cancer display high levels of Ho1 and Bach1. Bach1 transcriptional signature is associated with poor survival and metastasis in lung cancer patients. We propose that Nrf2 activates a metastatic program by inhibiting the heme- and Fbxo22-mediated degradation of Bach1, and that Ho1 inhibitors represent an effective therapeutic strategy to prevent lung cancer metastasis.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Neoplasias Pulmonares/patología , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/antagonistas & inhibidores , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Línea Celular Tumoral , Movimiento Celular , Proteínas F-Box/antagonistas & inhibidores , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Femenino , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Estimación de Kaplan-Meier , Proteína 1 Asociada A ECH Tipo Kelch/antagonistas & inhibidores , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Metástasis de la Neoplasia , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Activación Transcripcional
18.
Immunity ; 57(4): 840-842, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38599176

RESUMEN

Stress hormones can contribute to cancer progression, but how immune cells play a role in this process is unclear. In a recent study in Cancer Cell, He et al. showed that glucocorticoids potentiate metastasis by skewing neutrophils toward pro-tumorigenic functions.


Asunto(s)
Neoplasias , Neutrófilos , Humanos , Neoplasias/patología , Microambiente Tumoral , Metástasis de la Neoplasia/patología
19.
Cell ; 175(3): 751-765.e16, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30318143

RESUMEN

We examined how the immune microenvironment molds tumor evolution at different metastatic organs in a longitudinal dataset of colorectal cancer. Through multiplexed analyses, we showed that clonal evolution patterns during metastatic progression depend on the immune contexture at the metastatic site. Genetic evidence of neoantigen depletion was observed in the sites with high Immunoscore and spatial proximity between Ki67+ tumor cells and CD3+ cells. The immunoedited tumor clones were eliminated and did not recur, while progressing clones were immune privileged, despite the presence of tumor-infiltrating lymphocytes. Characterization of immune-privileged metastases revealed tumor-intrinsic and tumor-extrinsic mechanisms of escape. The lowest recurrence risk was associated with high Immunoscore, occurrence of immunoediting, and low tumor burden. We propose a parallel selection model of metastatic progression, where branched evolution could be traced back to immune-escaping clones. The findings could inform the understanding of cancer dissemination and the development of immunotherapeutics.


Asunto(s)
Infiltración Leucémica/inmunología , Modelos Estadísticos , Neoplasias/inmunología , Carga Tumoral/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/patología , Microambiente Tumoral/inmunología
20.
Cell ; 173(2): 338-354.e15, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29625051

RESUMEN

Cancer progression involves the gradual loss of a differentiated phenotype and acquisition of progenitor and stem-cell-like features. Here, we provide novel stemness indices for assessing the degree of oncogenic dedifferentiation. We used an innovative one-class logistic regression (OCLR) machine-learning algorithm to extract transcriptomic and epigenetic feature sets derived from non-transformed pluripotent stem cells and their differentiated progeny. Using OCLR, we were able to identify previously undiscovered biological mechanisms associated with the dedifferentiated oncogenic state. Analyses of the tumor microenvironment revealed unanticipated correlation of cancer stemness with immune checkpoint expression and infiltrating immune cells. We found that the dedifferentiated oncogenic phenotype was generally most prominent in metastatic tumors. Application of our stemness indices to single-cell data revealed patterns of intra-tumor molecular heterogeneity. Finally, the indices allowed for the identification of novel targets and possible targeted therapies aimed at tumor differentiation.


Asunto(s)
Desdiferenciación Celular/genética , Aprendizaje Automático , Neoplasias/patología , Carcinogénesis , Metilación de ADN , Bases de Datos Genéticas , Epigénesis Genética , Humanos , MicroARNs/metabolismo , Metástasis de la Neoplasia , Neoplasias/genética , Células Madre/citología , Células Madre/metabolismo , Transcriptoma , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA