Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(4): 644-658, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38503922

RESUMEN

The organization of immune cells in human tumors is not well understood. Immunogenic tumors harbor spatially localized multicellular 'immunity hubs' defined by expression of the T cell-attracting chemokines CXCL10/CXCL11 and abundant T cells. Here, we examined immunity hubs in human pre-immunotherapy lung cancer specimens and found an association with beneficial response to PD-1 blockade. Critically, we discovered the stem-immunity hub, a subtype of immunity hub strongly associated with favorable PD-1-blockade outcome. This hub is distinct from mature tertiary lymphoid structures and is enriched for stem-like TCF7+PD-1+CD8+ T cells, activated CCR7+LAMP3+ dendritic cells and CCL19+ fibroblasts as well as chemokines that organize these cells. Within the stem-immunity hub, we find preferential interactions between CXCL10+ macrophages and TCF7-CD8+ T cells as well as between mature regulatory dendritic cells and TCF7+CD4+ and regulatory T cells. These results provide a picture of the spatial organization of the human intratumoral immune response and its relevance to patient immunotherapy outcomes.


Asunto(s)
Neoplasias Pulmonares , Humanos , Linfocitos T CD8-positivos , Receptor de Muerte Celular Programada 1 , Quimiocinas/metabolismo , Inmunoterapia/métodos , Microambiente Tumoral
2.
Cell ; 184(26): 6262-6280.e26, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34910928

RESUMEN

Colorectal cancers (CRCs) arise from precursor polyps whose cellular origins, molecular heterogeneity, and immunogenic potential may reveal diagnostic and therapeutic insights when analyzed at high resolution. We present a single-cell transcriptomic and imaging atlas of the two most common human colorectal polyps, conventional adenomas and serrated polyps, and their resulting CRC counterparts. Integrative analysis of 128 datasets from 62 participants reveals adenomas arise from WNT-driven expansion of stem cells, while serrated polyps derive from differentiated cells through gastric metaplasia. Metaplasia-associated damage is coupled to a cytotoxic immune microenvironment preceding hypermutation, driven partly by antigen-presentation differences associated with tumor cell-differentiation status. Microsatellite unstable CRCs contain distinct non-metaplastic regions where tumor cells acquire stem cell properties and cytotoxic immune cells are depleted. Our multi-omic atlas provides insights into malignant progression of colorectal polyps and their microenvironment, serving as a framework for precision surveillance and prevention of CRC.


Asunto(s)
Pólipos del Colon/patología , Neoplasias Colorrectales/patología , Microambiente Tumoral , Inmunidad Adaptativa , Adenoma/genética , Adenoma/patología , Adulto , Anciano , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Muerte Celular , Diferenciación Celular , Pólipos del Colon/genética , Pólipos del Colon/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Heterogeneidad Genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mutación/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , RNA-Seq , Reproducibilidad de los Resultados , Análisis de la Célula Individual , Microambiente Tumoral/inmunología
3.
Cell ; 182(3): 655-671.e22, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32603654

RESUMEN

Checkpoint blockade with antibodies specific for the PD-1 and CTLA-4 inhibitory receptors can induce durable responses in a wide range of human cancers. However, the immunological mechanisms responsible for severe inflammatory side effects remain poorly understood. Here we report a comprehensive single-cell analysis of immune cell populations in colitis, a common and severe side effect of checkpoint blockade. We observed a striking accumulation of CD8 T cells with highly cytotoxic and proliferative states and no evidence of regulatory T cell depletion. T cell receptor (TCR) sequence analysis demonstrated that a substantial fraction of colitis-associated CD8 T cells originated from tissue-resident populations, explaining the frequently early onset of colitis symptoms following treatment initiation. Our analysis also identified cytokines, chemokines, and surface receptors that could serve as therapeutic targets for colitis and potentially other inflammatory side effects of checkpoint blockade.


Asunto(s)
Linfocitos T CD8-positivos/citología , Antígeno CTLA-4/inmunología , Colitis/metabolismo , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inmunoterapia/efectos adversos , Células Mieloides/metabolismo , Receptores de Quimiocina/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Antígeno CTLA-4/metabolismo , Quimiocinas/metabolismo , Colitis/tratamiento farmacológico , Colitis/genética , Colitis/inmunología , Citocinas/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Melanoma/genética , Melanoma/inmunología , Melanoma/metabolismo , Familia de Multigenes , Células Mieloides/citología , RNA-Seq , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores CXCR3/genética , Receptores CXCR3/metabolismo , Receptores CXCR6/genética , Receptores CXCR6/metabolismo , Receptores de Quimiocina/genética , Análisis de la Célula Individual , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo
4.
Cell ; 182(6): 1606-1622.e23, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32888429

RESUMEN

The enteric nervous system (ENS) coordinates diverse functions in the intestine but has eluded comprehensive molecular characterization because of the rarity and diversity of cells. Here we develop two methods to profile the ENS of adult mice and humans at single-cell resolution: RAISIN RNA-seq for profiling intact nuclei with ribosome-bound mRNA and MIRACL-seq for label-free enrichment of rare cell types by droplet-based profiling. The 1,187,535 nuclei in our mouse atlas include 5,068 neurons from the ileum and colon, revealing extraordinary neuron diversity. We highlight circadian expression changes in enteric neurons, show that disease-related genes are dysregulated with aging, and identify differences between the ileum and proximal/distal colon. In humans, we profile 436,202 nuclei, recovering 1,445 neurons, and identify conserved and species-specific transcriptional programs and putative neuro-epithelial, neuro-stromal, and neuro-immune interactions. The human ENS expresses risk genes for neuropathic, inflammatory, and extra-intestinal diseases, suggesting neuronal contributions to disease.


Asunto(s)
Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Neuronas/metabolismo , Cuerpos de Nissl/metabolismo , ARN Mensajero/metabolismo , Análisis de la Célula Individual/métodos , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Relojes Circadianos/genética , Colon/citología , Colon/metabolismo , Retículo Endoplásmico Rugoso/genética , Retículo Endoplásmico Rugoso/metabolismo , Retículo Endoplásmico Rugoso/ultraestructura , Células Epiteliales/metabolismo , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Íleon/citología , Íleon/metabolismo , Inflamación/genética , Inflamación/metabolismo , Enfermedades Intestinales/genética , Enfermedades Intestinales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Cuerpos de Nissl/genética , Cuerpos de Nissl/ultraestructura , ARN Mensajero/genética , RNA-Seq , Ribosomas/metabolismo , Ribosomas/ultraestructura , Células del Estroma/metabolismo
5.
Nat Immunol ; 23(10): 1495-1506, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36151395

RESUMEN

The immune system can eliminate tumors, but checkpoints enable immune escape. Here, we identify immune evasion mechanisms using genome-scale in vivo CRISPR screens across cancer models treated with immune checkpoint blockade (ICB). We identify immune evasion genes and important immune inhibitory checkpoints conserved across cancers, including the non-classical major histocompatibility complex class I (MHC class I) molecule Qa-1b/HLA-E. Surprisingly, loss of tumor interferon-γ (IFNγ) signaling sensitizes many models to immunity. The immune inhibitory effects of tumor IFN sensing are mediated through two mechanisms. First, tumor upregulation of classical MHC class I inhibits natural killer cells. Second, IFN-induced expression of Qa-1b inhibits CD8+ T cells via the NKG2A/CD94 receptor, which is induced by ICB. Finally, we show that strong IFN signatures are associated with poor response to ICB in individuals with renal cell carcinoma or melanoma. This study reveals that IFN-mediated upregulation of classical and non-classical MHC class I inhibitory checkpoints can facilitate immune escape.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico , Evasión Inmune , Interferón gamma/genética , Interferón gamma/metabolismo , Subfamília C de Receptores Similares a Lectina de Células NK
6.
Cell ; 176(6): 1325-1339.e22, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30827679

RESUMEN

Lineage tracing provides key insights into the fate of individual cells in complex organisms. Although effective genetic labeling approaches are available in model systems, in humans, most approaches require detection of nuclear somatic mutations, which have high error rates, limited scale, and do not capture cell state information. Here, we show that somatic mutations in mtDNA can be tracked by single-cell RNA or assay for transposase accessible chromatin (ATAC) sequencing. We leverage somatic mtDNA mutations as natural genetic barcodes and demonstrate their utility as highly accurate clonal markers to infer cellular relationships. We track native human cells both in vitro and in vivo and relate clonal dynamics to gene expression and chromatin accessibility. Our approach should allow clonal tracking at a 1,000-fold greater scale than with nuclear genome sequencing, with simultaneous information on cell state, opening the way to chart cellular dynamics in human health and disease.


Asunto(s)
ADN Mitocondrial/genética , Mitocondrias/genética , Secuencia de Bases , Linaje de la Célula , Cromatina , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Genómica/métodos , Células HEK293 , Células Madre Hematopoyéticas/fisiología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Mutación , Análisis de la Célula Individual , Transposasas
7.
Cell ; 174(4): 843-855.e19, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30017245

RESUMEN

Many patients with advanced cancers achieve dramatic responses to a panoply of therapeutics yet retain minimal residual disease (MRD), which ultimately results in relapse. To gain insights into the biology of MRD, we applied single-cell RNA sequencing to malignant cells isolated from BRAF mutant patient-derived xenograft melanoma cohorts exposed to concurrent RAF/MEK-inhibition. We identified distinct drug-tolerant transcriptional states, varying combinations of which co-occurred within MRDs from PDXs and biopsies of patients on treatment. One of these exhibited a neural crest stem cell (NCSC) transcriptional program largely driven by the nuclear receptor RXRG. An RXR antagonist mitigated accumulation of NCSCs in MRD and delayed the development of resistance. These data identify NCSCs as key drivers of resistance and illustrate the therapeutic potential of MRD-directed therapy. They also highlight how gene regulatory network architecture reprogramming may be therapeutically exploited to limit cellular heterogeneity, a key driver of disease progression and therapy resistance.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Melanoma/tratamiento farmacológico , Neoplasia Residual/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Receptor gamma X Retinoide/antagonistas & inhibidores , Animales , Biomarcadores de Tumor , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/genética , Masculino , Melanoma/metabolismo , Melanoma/patología , Ratones SCID , Mutación , Neoplasia Residual/metabolismo , Neoplasia Residual/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cell ; 175(4): 998-1013.e20, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388456

RESUMEN

Treatment of cancer has been revolutionized by immune checkpoint blockade therapies. Despite the high rate of response in advanced melanoma, the majority of patients succumb to disease. To identify factors associated with success or failure of checkpoint therapy, we profiled transcriptomes of 16,291 individual immune cells from 48 tumor samples of melanoma patients treated with checkpoint inhibitors. Two distinct states of CD8+ T cells were defined by clustering and associated with patient tumor regression or progression. A single transcription factor, TCF7, was visualized within CD8+ T cells in fixed tumor samples and predicted positive clinical outcome in an independent cohort of checkpoint-treated patients. We delineated the epigenetic landscape and clonality of these T cell states and demonstrated enhanced antitumor immunity by targeting novel combinations of factors in exhausted cells. Our study of immune cell transcriptomes from tumors demonstrates a strategy for identifying predictors, mechanisms, and targets for enhancing checkpoint immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunoterapia/métodos , Melanoma/inmunología , Transcriptoma , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Antígenos CD/inmunología , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Apirasa/antagonistas & inhibidores , Apirasa/inmunología , Línea Celular Tumoral , Humanos , Antígenos Comunes de Leucocito/antagonistas & inhibidores , Antígenos Comunes de Leucocito/inmunología , Melanoma/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor 1 de Transcripción de Linfocitos T/metabolismo
9.
Cell ; 175(4): 984-997.e24, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388455

RESUMEN

Immune checkpoint inhibitors (ICIs) produce durable responses in some melanoma patients, but many patients derive no clinical benefit, and the molecular underpinnings of such resistance remain elusive. Here, we leveraged single-cell RNA sequencing (scRNA-seq) from 33 melanoma tumors and computational analyses to interrogate malignant cell states that promote immune evasion. We identified a resistance program expressed by malignant cells that is associated with T cell exclusion and immune evasion. The program is expressed prior to immunotherapy, characterizes cold niches in situ, and predicts clinical responses to anti-PD-1 therapy in an independent cohort of 112 melanoma patients. CDK4/6-inhibition represses this program in individual malignant cells, induces senescence, and reduces melanoma tumor outgrowth in mouse models in vivo when given in combination with immunotherapy. Our study provides a high-resolution landscape of ICI-resistant cell states, identifies clinically predictive signatures, and suggests new therapeutic strategies to overcome immunotherapy resistance.


Asunto(s)
Antineoplásicos/uso terapéutico , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Melanoma/inmunología , Inhibidores de Proteínas Quinasas/uso terapéutico , Linfocitos T/inmunología , Escape del Tumor , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Femenino , Humanos , Inmunoterapia/métodos , Masculino , Melanoma/tratamiento farmacológico , Melanoma/terapia , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología
11.
Nat Immunol ; 20(9): 1231-1243, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31358999

RESUMEN

Understanding resistance to antibody to programmed cell death protein 1 (PD-1; anti-PD-1) is crucial for the development of reversal strategies. In anti-PD-1-resistant models, simultaneous anti-PD-1 and vaccine therapy reversed resistance, while PD-1 blockade before antigen priming abolished therapeutic outcomes. This was due to induction of dysfunctional PD-1+CD38hi CD8+ cells by PD-1 blockade in suboptimally primed CD8 cell conditions induced by tumors. This results in erroneous T cell receptor signaling and unresponsiveness to antigenic restimulation. On the other hand, PD-1 blockade of optimally primed CD8 cells prevented the induction of dysfunctional CD8 cells, reversing resistance. Depleting PD-1+CD38hi CD8+ cells enhanced therapeutic outcomes. Furthermore, non-responding patients showed more PD-1+CD38+CD8+ cells in tumor and blood than responders. In conclusion, the status of CD8+ T cell priming is a major contributor to anti-PD-1 therapeutic resistance. PD-1 blockade in unprimed or suboptimally primed CD8 cells induces resistance through the induction of PD-1+CD38hi CD8+ cells that is reversed by optimal priming. PD-1+CD38hi CD8+ cells serve as a predictive and therapeutic biomarker for anti-PD-1 treatment. Sequencing of anti-PD-1 and vaccine is crucial for successful therapy.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Linfocitos T CD8-positivos/inmunología , Resistencia a Antineoplásicos/inmunología , Glicoproteínas de Membrana/metabolismo , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , ADP-Ribosil Ciclasa 1/genética , Animales , Anticuerpos/inmunología , Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Inmunoterapia/métodos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Microambiente Tumoral/inmunología
13.
Immunity ; 50(6): 1498-1512.e5, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31097342

RESUMEN

Despite compelling rates of durable clinical responses to programmed cell death-1 (PD-1) blockade, advances are needed to extend these benefits to resistant tumors. We found that tumor-bearing mice deficient in the chemokine receptor CXCR3 responded poorly to anti-PD-1 treatment. CXCR3 and its ligand CXCL9 were critical for a productive CD8+ T cell response in tumor-bearing mice treated with anti-PD-1 but were not required for the infiltration of CD8+ T cells into tumors. The anti-PD-1-induced anti-tumor response was facilitated by CXCL9 production from intratumoral CD103+ dendritic cells, suggesting that CXCR3 facilitates dendritic cell-T cell interactions within the tumor microenvironment. CXCR3 ligands in murine tumors and in plasma of melanoma patients were an indicator of clinical response to anti-PD-1, and their induction in non-responsive murine tumors promoted responsiveness to anti-PD-1. Our data suggest that the CXCR3 chemokine system is a biomarker for sensitivity to PD-1 blockade and that augmenting the intratumoral function of this chemokine system could improve clinical outcomes.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Inmunomodulación/efectos de los fármacos , Neoplasias/inmunología , Neoplasias/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Receptores CXCR3/metabolismo , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Epigénesis Genética , Humanos , Activación de Linfocitos , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Nature ; 605(7910): 532-538, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35508657

RESUMEN

Within the tumour microenvironment, CD4+ T cells can promote or suppress antitumour responses through the recognition of antigens presented by human leukocyte antigen (HLA) class II molecules1,2, but how cancers co-opt these physiologic processes to achieve immune evasion remains incompletely understood. Here we performed in-depth analysis of the phenotype and tumour specificity of CD4+ T cells infiltrating human melanoma specimens, finding that exhausted cytotoxic CD4+ T cells could be directly induced by melanoma cells through recognition of HLA class II-restricted neoantigens, and also HLA class I-restricted tumour-associated antigens. CD4+ T regulatory (TReg) cells could be indirectly elicited through presentation of tumour antigens via antigen-presenting cells. Notably, numerous tumour-reactive CD4+ TReg clones were stimulated directly by HLA class II-positive melanoma and demonstrated specificity for melanoma neoantigens. This phenomenon was observed in the presence of an extremely high tumour neoantigen load, which we confirmed to be associated with HLA class II positivity through the analysis of 116 melanoma specimens. Our data reveal the landscape of infiltrating CD4+ T cells in melanoma and point to the presentation of HLA class II-restricted neoantigens and direct engagement of immunosuppressive CD4+ TReg cells as a mechanism of immune evasion that is favoured in HLA class II-positive melanoma.


Asunto(s)
Antígenos de Neoplasias , Linfocitos T CD4-Positivos , Melanoma , Neoplasias Cutáneas , Células Presentadoras de Antígenos , Antígenos de Neoplasias/inmunología , Antígenos HLA , Humanos , Melanoma/inmunología , Fenotipo , Neoplasias Cutáneas/inmunología , Células Tumorales Cultivadas , Microambiente Tumoral
15.
Nature ; 596(7870): 119-125, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34290406

RESUMEN

Interactions between T cell receptors (TCRs) and their cognate tumour antigens are central to antitumour immune responses1-3; however, the relationship between phenotypic characteristics and TCR properties is not well elucidated. Here we show, by linking the antigenic specificity of TCRs and the cellular phenotype of melanoma-infiltrating lymphocytes at single-cell resolution, that tumour specificity shapes the expression state of intratumoural CD8+ T cells. Non-tumour-reactive T cells were enriched for viral specificities and exhibited a non-exhausted memory phenotype, whereas melanoma-reactive lymphocytes predominantly displayed an exhausted state that encompassed diverse levels of differentiation but rarely acquired memory properties. These exhausted phenotypes were observed both among clonotypes specific for public overexpressed melanoma antigens (shared across different tumours) or personal neoantigens (specific for each tumour). The recognition of such tumour antigens was provided by TCRs with avidities inversely related to the abundance of cognate targets in melanoma cells and proportional to the binding affinity of peptide-human leukocyte antigen (HLA) complexes. The persistence of TCR clonotypes in peripheral blood was negatively affected by the level of intratumoural exhaustion, and increased in patients with a poor response to immune checkpoint blockade, consistent with chronic stimulation mediated by residual tumour antigens. By revealing how the quality and quantity of tumour antigens drive the features of T cell responses within the tumour microenvironment, we gain insights into the properties of the anti-melanoma TCR repertoire.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Melanoma/inmunología , Especificidad por Sustrato/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Conjuntos de Datos como Asunto , Regulación de la Expresión Génica , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/sangre , Fenotipo , Receptores de Antígenos de Linfocitos T/inmunología , Análisis de la Célula Individual , Transcriptoma/genética , Microambiente Tumoral
17.
Ann Surg Oncol ; 31(4): 2713-2726, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38158497

RESUMEN

BACKGROUND: In the current era of effective adjuvant therapies and de-escalation of surgery, distinguishing which patients with high-risk stage II melanoma are at increased risk of recurrence after excision of the primary lesion is essential to determining appropriate treatment and surveillance plans. METHODS: A single-center retrospective study analyzed patients with stage IIB or IIC melanoma. Demographic and tumor data were collected, and genomic analysis of formalin-fixed, paraffin-embedded tissue samples was performed via an internal next-generation sequencing (NGS) platform (SNaPshot). The end points examined were relapse-free survival (RFS), distant metastasis-free survival (DMFS), overall survival (OS), and melanoma-specific survival (MSS). Uni- and multivariable Cox regressions were performed to calculate the hazard ratios. RESULTS: The study included 92 patients with a median age of 69 years and a male/female ratio of 2:1. A Breslow depth greater than 4 mm, a higher mitotic rate, an advanced T stage, and a KIT mutation had a negative impact on RFS. A primary lesion in the head and neck, a mitotic rate exceeding 10 mitoses per mm2, a CDH1 mutation, or a KIT mutation was significantly associated with a shorter DMFS. Overall survival was significantly lower with older age at diagnosis and a higher mitotic rate. An older age at diagnosis also had a negative impact on MSS. CONCLUSION: Traditional histopathologic factors and specific tumor mutations displayed a significant correlation with disease recurrence and survival for patients with high-risk stage II melanoma. This study supported the use of genomic testing of high-risk stage II melanomas for prognostic prediction and risk stratification.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Femenino , Masculino , Anciano , Melanoma/genética , Melanoma/cirugía , Melanoma/patología , Estudios Retrospectivos , Estadificación de Neoplasias , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/cirugía , Recurrencia Local de Neoplasia/epidemiología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/cirugía , Neoplasias Cutáneas/patología , Pronóstico , Tasa de Supervivencia
18.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34497125

RESUMEN

Absolute quantification measurements (copies per cell) of peptide major histocompatibility complex (pMHC) antigens are necessary to inform targeted immunotherapy drug design; however, existing methods for absolute quantification have critical limitations. Here, we present a platform termed SureQuant-IsoMHC, utilizing a series of pMHC isotopologues and internal standard-triggered targeted mass spectrometry to generate an embedded multipoint calibration curve to determine endogenous pMHC concentrations for a panel of 18 tumor antigens. We apply SureQuant-IsoMHC to measure changes in expression of our target panel in a melanoma cell line treated with a MEK inhibitor and translate this approach to estimate antigen concentrations in melanoma tumor biopsies.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/análisis , Bencimidazoles/farmacología , Antígenos de Histocompatibilidad Clase I/inmunología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Melanoma/inmunología , Presentación de Antígeno/efectos de los fármacos , Antígenos de Neoplasias/efectos de los fármacos , Antígenos de Neoplasias/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoterapia , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Células Tumorales Cultivadas
19.
Ann Surg Oncol ; 30(8): 5013-5014, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37225835

RESUMEN

The role of surgery for patients with stage IV melanoma is rapidly evolving. In the past, limited treatment options were available and surgery was offered to carefully selected patients. Currently, in the era of effective immunotherapy, the role of surgery is still being defined. The present study examines outcomes for patients with stage IV melanoma receiving immunotherapy and surgery. Future studies will help to better identify which patients should receive surgery and when it should be performed in the setting of increasingly available therapeutic modalities for patients with stage IV melanoma.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Melanoma/cirugía , Inmunoterapia , Neoplasias Cutáneas/cirugía
20.
Ann Surg Oncol ; 30(8): 5005-5012, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37121988

RESUMEN

BACKGROUND: The benefit of surgery for patients with stage IV melanoma in the modern era of effective immunotherapy is unclear. This study aimed to evaluate trends and outcomes after surgical resection of stage IV melanoma in the modern immunotherapy era. METHODS: Patients with stage IV melanoma who received surgery, immunotherapy, or both from 2012 to 2017 were identified from the National Cancer Database (NCDB). Demographics, facility-level characteristics, and use of immunotherapy were compared between patients who received surgery and those who did not. Multivariate Poisson regression modeling, Kaplan-Meier survival analysis, and Cox regression analysis were performed. RESULTS: The study identified 9800 patients with stage IV melanoma, and 2160 of these patients (22 %) underwent surgery. The patients who received surgery were more likely to be younger (P < 0.001), to have private insurance (P < 0.001), to have a higher median income (P = 0.008), and to receive treatment at academic/research programs (P < 0.001), whereas they were less likely to receive immunotherapy (33.7 % vs 36.6 %; P = 0.013). The patients who received immunotherapy had a lower likelihood of undergoing surgery (relative risk [RR], 0.82; 95 % confidence interval [CI[, 0.75-0.88; P < 0.001). The patients who received both surgery and immunotherapy had a better overall survival rate (hazard ratio [HR], 0.41; 95 % CI, 0.36-0.46; P < 0.01) than the patients who received neither immunotherapy nor surgery. CONCLUSIONS: The use of immunotherapy was associated with a lower use of surgery for patients with stage IV melanoma. The patients with stage IV disease who received both surgery and immunotherapy had the highest overall survival rates.


Asunto(s)
Melanoma , Humanos , Melanoma/cirugía , Inmunoterapia , Modelos de Riesgos Proporcionales , Terapia Combinada , Análisis de Regresión , Estadificación de Neoplasias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA