Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nature ; 577(7791): 556-560, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31942077

RESUMEN

Soft-tissue sarcomas represent a heterogeneous group of cancer, with more than 50 histological subtypes1,2. The clinical presentation of patients with different subtypes is often atypical, and responses to therapies such as immune checkpoint blockade vary widely3,4. To explain this clinical variability, here we study gene expression profiles in 608 tumours across subtypes of soft-tissue sarcoma. We establish an immune-based classification on the basis of the composition of the tumour microenvironment and identify five distinct phenotypes: immune-low (A and B), immune-high (D and E), and highly vascularized (C) groups. In situ analysis of an independent validation cohort shows that class E was characterized by the presence of tertiary lymphoid structures that contain T cells and follicular dendritic cells and are particularly rich in B cells. B cells are the strongest prognostic factor even in the context of high or low CD8+ T cells and cytotoxic contents. The class-E group demonstrated improved survival and a high response rate to PD1 blockade with pembrolizumab in a phase 2 clinical trial. Together, this work confirms the immune subtypes in patients with soft-tissue sarcoma, and unravels the potential of B-cell-rich tertiary lymphoid structures to guide clinical decision-making and treatments, which could have broader applications in other diseases.


Asunto(s)
Linfocitos B/inmunología , Inmunoterapia , Sarcoma/tratamiento farmacológico , Sarcoma/inmunología , Estructuras Linfoides Terciarias/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Estudios de Cohortes , Células Dendríticas Foliculares/inmunología , Humanos , Mutación , Fenotipo , Pronóstico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Reproducibilidad de los Resultados , Sarcoma/clasificación , Sarcoma/patología , Tasa de Supervivencia , Microambiente Tumoral
2.
Blood ; 135(5): 360-370, 2020 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-31774495

RESUMEN

The oncogenic events involved in breast implant-associated anaplastic large cell lymphoma (BI-ALCL) remain elusive. To clarify this point, we have characterized the genomic landscape of 34 BI-ALCLs (15 tumor and 19 in situ subtypes) collected from 54 BI-ALCL patients diagnosed through the French Lymphopath network. Whole-exome sequencing (n = 22, with paired tumor/germline DNA) and/or targeted deep sequencing (n = 24) showed recurrent mutations of epigenetic modifiers in 74% of cases, involving notably KMT2C (26%), KMT2D (9%), CHD2 (15%), and CREBBP (15%). KMT2D and KMT2C mutations correlated with a loss of H3K4 mono- and trimethylation by immunohistochemistry. Twenty cases (59%) showed mutations in ≥1 member of the JAK/STAT pathway, including STAT3 (38%), JAK1 (18%), and STAT5B (3%), and in negative regulators, including SOCS3 (6%), SOCS1 (3%), and PTPN1 (3%). These mutations were more frequent in tumor-type samples than in situ samples (P = .038). All BI-ALCLs expressed pSTAT3, regardless of the mutational status of genes in the JAK/STAT pathway. Mutations in the EOMES gene (12%) involved in lymphocyte development, PI3K-AKT/mTOR (6%), and loss-of-function mutations in TP53 (12%) were also identified. Copy-number aberration (CNA) analysis identified recurrent alterations, including gains on chromosomes 2, 9p, 12p, and 21 and losses on 4q, 8p, 15, 16, and 20. Regions of CNA encompassed genes involved in the JAK/STAT pathway and epigenetic regulators. Our results show that the BI-ALCL genomic landscape is characterized by not only JAK/STAT activating mutations but also loss-of-function alterations of epigenetic modifiers.


Asunto(s)
Implantes de Mama/efectos adversos , Epigénesis Genética , Quinasas Janus/metabolismo , Linfoma Anaplásico de Células Grandes/etiología , Linfoma Anaplásico de Células Grandes/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Adulto , Anciano , Anciano de 80 o más Años , Variaciones en el Número de Copia de ADN , Femenino , Genoma Humano , Humanos , Linfoma Anaplásico de Células Grandes/patología , Persona de Mediana Edad , Mutación/genética
3.
Br J Cancer ; 120(8): 815-818, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30862951

RESUMEN

In colorectal cancer (CRC), T-cell checkpoint blockade is only effective in patients diagnosed with mismatch repair-deficient (MMR-d) cancers. However, defects in Human Leukocyte Antigen (HLA) class I expression were reported to occur in most MMR-d CRCs, which would preclude antigen presentation in these tumours, considered essential for the clinical activity of this immunotherapeutic modality. We revisited this paradox by characterising HLA class I expression in two independent cohorts of CRC. We determined that loss of HLA class I expression occurred in the majority (73-78%) of MMR-d cases. This phenotype was rare in CRC liver metastases, irrespective of MMR status, whereas weak, inducible expression of HLA class I molecules was frequent in liver lesions. We propose that HLA class I is an important determinant of metastatic homing in CRCs. This observation is paramount to understand CRC carcinogenesis and for the application of immunotherapies in the metastatic setting.


Asunto(s)
Neoplasias Colorrectales/terapia , Genes MHC Clase I/genética , Inmunoterapia , Linfocitos T/inmunología , Presentación de Antígeno/inmunología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Carcinogénesis/genética , Carcinogénesis/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Genes cdc/efectos de los fármacos , Genes cdc/inmunología , Humanos , Hígado/metabolismo , Hígado/patología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/inmunología
4.
Cancer Immunol Immunother ; 68(1): 109-120, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30315348

RESUMEN

IL-1 family cytokines play a dual role in the gut, with different family members contributing either protective or pathogenic effects. IL-36γ is an IL-1 family cytokine involved in polarizing type-1 immune responses. However, its function in the gut, including in colorectal cancer pathogenesis, is not well appreciated. In a murine model of colon carcinoma, IL-36γ controls tertiary lymphoid structure formation and promotes a type-1 immune response concurrently with a decrease in expression of immune checkpoint molecules in the tumor microenvironment. Here, we demonstrate that IL-36γ plays a similar role in driving a pro-inflammatory phenotype in human colorectal cancer. We analyzed a cohort of 33 primary colorectal carcinoma tumors using imaging, flow cytometry, and transcriptomics to determine the pattern and role of IL-36γ expression in this disease. In the colorectal tumor microenvironment, we observed IL-36γ to be predominantly expressed by M1 macrophages and cells of the vasculature, including smooth muscle cells and high endothelial venules. This pattern of IL-36γ expression is associated with a CD4+ central memory T cell infiltrate and an increased density of B cells in tertiary lymphoid structures, as well as with markers of fibrosis. Conversely, expression of the antagonist to IL-36 signaling, IL-1F5, was associated with intratumoral expression of checkpoint molecules, including PD-1, PD-L1, and CTLA4, which can suppress the immune response. These data support a role for IL-36γ in the physiologic immune response to colorectal cancer by sustaining inflammation within the tumor microenvironment.


Asunto(s)
Neoplasias Colorrectales/inmunología , Inflamación/inmunología , Interleucina-1/inmunología , Estructuras Linfoides Terciarias/inmunología , Anciano , Anciano de 80 o más Años , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Interleucina-1/metabolismo , Masculino , Persona de Mediana Edad , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
6.
Methods Mol Biol ; 2227: 191-203, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33847943

RESUMEN

Tumors contain a complement rich microenvironment in which all cell types (e.g., tumor cells and stromal cells) are able to produce different proteins. We developed immunohistochemistry (IHC) assays allowing to identify on paraffin embedded tumor sections, not only the complement producing cells but also the complement activation fragments which result from activation of complement cascade within the tumor. The local production of complement can be detected by cytoplasmic staining, whereas the activation fragments are localized at the surface of the cells. There is a high heterogeneity of the staining within tumors but also between patients. Semi-quantification of the staining in large cohorts of patients allows to investigate the prognostic impact of the local complement production and activation. Here we explain the staining process for C1q, C4, and C3 in human paraffin-embedded tumor sections by immunofluorescence and immunohistochemistry.


Asunto(s)
Proteínas del Sistema Complemento/metabolismo , Técnica del Anticuerpo Fluorescente/métodos , Inmunohistoquímica/métodos , Neoplasias/metabolismo , Animales , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Proteínas del Sistema Complemento/análisis , Cabras , Humanos , Ratones , Neoplasias/diagnóstico , Neoplasias/inmunología , Neoplasias/patología , Pronóstico , Conejos , Microambiente Tumoral/inmunología
7.
Clin Cancer Res ; 26(16): 4381-4389, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32269054

RESUMEN

PURPOSE: The impact of tertiary lymphoid structures (TLS) in hepatocellular carcinoma (HCC) progression is being extensively investigated. However, their presence during the early steps of human liver carcinogenesis remains unknown. We thus aimed to determine whether TLS are induced in preneoplastic/early hepatic lesions (EHL), and whether they are associated with a particular immune profile. EXPERIMENTAL DESIGN: A series of 127 EHLs (low/high-grade dysplastic nodules, early HCC, and small and progressed HCC) was included in the study. TLSs were investigated by pathologic reviewing. Densities of immune cells were assessed using IHC. A subset of lesions was microdissected and gene expression profiling was performed with a custom NanoString panel. RESULTS: Compared with surrounding cirrhotic nodules, EHL of all stages displayed increased densities of T cells, B cells, and dendritic cells. Immature TLSs were identified in 24% of EHL. Gene expression profiling identified a subset of EHL with elevated mRNA levels of various cytokines involved in immune cells' recruitment and TLS induction. This subgroup of EHL also showed overexpression of genes related to T- and B-cells' activation and antigen presentation, as well as those related to immunosuppression and immune exhaustion. CONCLUSIONS: Local immune activation occurs in the very early steps of liver carcinogenesis; however, it may not be fully efficient and paradoxically favor immune evasion and progression to full-blown HCC. These results have implications for the development of anti-HCC chemopreventive strategies in cirrhotic patients.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Neoplasias Hepáticas/inmunología , Hígado/inmunología , Estructuras Linfoides Terciarias/genética , Anciano , Linfocitos B/inmunología , Biomarcadores de Tumor/inmunología , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Linfocitos T/inmunología , Estructuras Linfoides Terciarias/inmunología , Estructuras Linfoides Terciarias/patología
8.
Eur Urol Focus ; 5(2): 192-196, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-28753812

RESUMEN

Prostate cancer (PCa) patients with lymph node invasion at radical prostatectomy are at higher risk of tumor recurrence and receive immediate androgen deprivation therapy (ADT). While approximately 30% of these patients do not experience recurrence, others experience disease recurrence despite ADT, and currently no biomarkers can accurately identify them. We analyzed tumors from 51 patients with node-positive prostate cancer using immunohistochemistry to investigate whether expression of the immune checkpoint ligand PD-L1 by tumor cells or the density of CD8+ or CD20+ cells are associated with clinical progression. Patients with at least 1% PD-L1+ tumor cells had shorter metastasis-free survival than those with PD-L1- tumors (p=0.008, log-rank test). Univariate Cox regression showed that patients with PD-L1+ tumors had almost four times the risk of experiencing distant metastases than those with PD-L1- tumors (hazard ratio 3.90). In addition, we found that PD-L1 expression was significantly associated with CD8+ T-cell density, but not with CD20+ B-cell density. While these results need to be confirmed in larger studies, they show that PD-L1 and CD8 may be used as biomarkers for node-positive patients at high risk of progression. The study also provides a rationale for selecting patients with node-positive PCa who might benefit the most from adjuvant immunotherapies. PATIENT SUMMARY: None of the available biomarkers can identify node-positive prostate cancer that will recur after surgery. We found that expression of PD-L1 by tumor cells and a high density of CD8+ T cells in tumor are associated with a higher risk of clinical progression in men with node-positive prostate cancer.


Asunto(s)
Antígeno B7-H1/metabolismo , Biomarcadores de Tumor/metabolismo , Linfocitos T CD8-positivos/metabolismo , Neoplasias de la Próstata/patología , Adyuvantes Inmunológicos/uso terapéutico , Antagonistas de Andrógenos/uso terapéutico , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Humanos , Ganglios Linfáticos/patología , Metástasis Linfática , Masculino , Recurrencia Local de Neoplasia/epidemiología , Prostatectomía , Neoplasias de la Próstata/secundario , Neoplasias de la Próstata/cirugía , Estudios Retrospectivos
9.
Clin Cancer Res ; 25(24): 7351-7362, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31530631

RESUMEN

PURPOSE: The immunomodulatory activity of DNA hypomethylating agents (DHAs) suggests they may improve the effectiveness of cancer immunotherapies. The phase Ib NIBIT-M4 trial tested this hypothesis using the next-generation DHA guadecitabine combined with ipilimumab. PATIENTS AND METHODS: Patients with unresectable stage III/IV melanoma received escalating doses of guadecitabine 30, 45, or 60 mg/m2/day subcutaneously on days 1 to 5 every 3 weeks, and ipilimumab 3 mg/kg intravenously on day 1 every 3 weeks, starting 1 week after guadecitabine, for four cycles. Primary endpoints were safety, tolerability, and MTD of treatment; secondary were immune-related (ir) disease control rate (DCR) and objective response rate (ORR); and exploratory were changes in methylome, transcriptome, and immune contextures in sequential tumor biopsies, and pharmacokinetics. RESULTS: Nineteen patients were treated; 84% had grade 3/4 adverse events, and neither dose-limiting toxicities per protocol nor overlapping toxicities were observed. Ir-DCR and ir-ORR were 42% and 26%, respectively. Median CpG site methylation of tumor samples (n = 8) at week 4 (74.5%) and week 12 (75.5%) was significantly (P < 0.05) lower than at baseline (80.3%), with a median of 2,454 (week 4) and 4,131 (week 12) differentially expressed genes. Among the 136 pathways significantly (P < 0.05; Z score >2 or ←2) modulated by treatment, the most frequently activated were immune-related. Tumor immune contexture analysis (n = 11) demonstrated upregulation of HLA class I on melanoma cells, an increase in CD8+, PD-1+ T cells and in CD20+ B cells in posttreatment tumor cores. CONCLUSIONS: Treatment of guadecitabine combined with ipilimumab is safe and tolerable in advanced melanoma and has promising immunomodulatory and antitumor activity.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Metilación de ADN , Melanoma/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ipilimumab/administración & dosificación , Masculino , Dosis Máxima Tolerada , Melanoma/inmunología , Melanoma/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Seguridad del Paciente , Tasa de Supervivencia , Resultado del Tratamiento
10.
Cancer Immunol Res ; 7(7): 1091-1105, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31164356

RESUMEN

Clear-cell renal cell carcinoma (ccRCC) possesses an unmet medical need, particularly at the metastatic stage, when surgery is ineffective. Complement is a key factor in tissue inflammation, favoring cancer progression through the production of complement component 5a (C5a). However, the activation pathways that generate C5a in tumors remain obscure. By data mining, we identified ccRCC as a cancer type expressing concomitantly high expression of the components that are part of the classical complement pathway. To understand how the complement cascade is activated in ccRCC and impacts patients' clinical outcome, primary tumors from three patient cohorts (n = 106, 154, and 43), ccRCC cell lines, and tumor models in complement-deficient mice were used. High densities of cells producing classical complement pathway components C1q and C4 and the presence of C4 activation fragment deposits in primary tumors correlated with poor prognosis. The in situ orchestrated production of C1q by tumor-associated macrophages (TAM) and C1r, C1s, C4, and C3 by tumor cells associated with IgG deposits, led to C1 complex assembly, and complement activation. Accordingly, mice deficient in C1q, C4, or C3 displayed decreased tumor growth. However, the ccRCC tumors infiltrated with high densities of C1q-producing TAMs exhibited an immunosuppressed microenvironment, characterized by high expression of immune checkpoints (i.e., PD-1, Lag-3, PD-L1, and PD-L2). Our data have identified the classical complement pathway as a key inflammatory mechanism activated by the cooperation between tumor cells and TAMs, favoring cancer progression, and highlight potential therapeutic targets to restore an efficient immune reaction to cancer.


Asunto(s)
Carcinoma de Células Renales/patología , Complemento C1q/inmunología , Complemento C3/inmunología , Complemento C4/inmunología , Neoplasias Renales/patología , Macrófagos/inmunología , Microambiente Tumoral/inmunología , Animales , Apoptosis , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/metabolismo , Proliferación Celular , Activación de Complemento , Complemento C1q/metabolismo , Complemento C3/metabolismo , Complemento C4/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Factores Inmunológicos/metabolismo , Neoplasias Renales/inmunología , Neoplasias Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Estudios Retrospectivos , Tasa de Supervivencia , Células Tumorales Cultivadas
11.
Front Oncol ; 8: 390, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319963

RESUMEN

Tumors are formed by aggregates of cells of various origins including malignant, stromal and immune cells. The number of therapies targeting the microenvironment is increasing as the tumor microenvironment is more and more recognized as playing an essential role in tumor control. In the era of precision medicine, it is essential to precisely estimate the composition, organization and functionality of the individual patient tumor microenvironment and to find ways to therapeutically modulate it. To quantify the cell populations present in the tumor microenvironment, many tools are now available and the most recent approaches will be reviewed herein. We provide an overview of experimental and computational methodologies used to quantify tumor-associated cellular populations, including immunohistochemistry, flow and mass cytometry, bulk and single-cell transcriptomic approaches. We illustrate their respective contribution to characterize the microenvironment. We also discuss how these methods allow to guide therapeutic choices, in relation to the predictive value of some characteristics of the microenvironment.

12.
Clin Cancer Res ; 23(15): 4416-4428, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28213366

RESUMEN

Purpose: The efficacy of PD-1 checkpoint blockade as adjuvant therapy in localized clear cell renal cell carcinoma (ccRCC) is currently unknown. The identification of tumor microenvironment (TME) prognostic biomarkers in this setting may help define which patients could benefit from checkpoint blockade and uncover new therapeutic targets.Experimental Design: We performed multiparametric flow cytometric immunophenotypic analysis of T cells isolated from tumor tissue [tumor-infiltrating lymphocytes (TIL)], adjacent non-malignant renal tissue [renal-infiltrating lymphocytes (RIL)], and peripheral blood lymphocytes (PBL), in a cohort of patients (n = 40) with localized ccRCC. Immunophenotypic data were integrated with prognostic and histopathologic variables, T-cell receptor (TCR) repertoire analysis of sorted CD8+PD-1+ TILs, tumor mRNA expression, and digital quantitative immunohistochemistry.Results: On the basis of TIL phenotypic characterization, we identified three dominant immune profiles in localized ccRCC: (i) immune-regulated, characterized by polyclonal/poorly cytotoxic CD8+PD-1+Tim-3+Lag-3+ TILs and CD4+ICOS+ cells with a Treg phenotype (CD25+CD127-Foxp3+/Helios+GITR+), that developed in inflamed tumors with prominent infiltrations by dysfunctional dendritic cells and high PD-L1 expression; (ii) immune-activated, enriched in oligoclonal/cytotoxic CD8+PD-1+Tim-3+ TILs, that represented 22% of the tumors; and (iii) immune-silent, enriched in TILs exhibiting RIL-like phenotype, that represented 56% of patients in the cohort. Only immune-regulated tumors displayed aggressive histologic features, high risk of disease progression in the year following nephrectomy, and a CD8+PD-1+Tim-3+ and CD4+ICOS+ PBL phenotypic signature.Conclusions: In localized ccRCC, the infiltration with CD8+PD-1+Tim-3+Lag-3+ exhausted TILs and ICOS+ Treg identifies the patients with deleterious prognosis who could benefit from adjuvant therapy with TME-modulating agents and checkpoint blockade. This work also provides PBL phenotypic markers that could allow their identification. Clin Cancer Res; 23(15); 4416-28. ©2017 AACR.


Asunto(s)
Carcinoma de Células Renales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Pronóstico , Linfocitos T/inmunología , Anciano , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Células Dendríticas/inmunología , Células Dendríticas/patología , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunofenotipificación , Linfocitos Infiltrantes de Tumor/patología , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Linfocitos T/patología
13.
Clin Cancer Res ; 22(16): 4057-66, 2016 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26994146

RESUMEN

PURPOSE: The tumor microenvironment is formed by many distinct and interacting cell populations, and its composition may predict patients' prognosis and response to therapies. Colorectal cancer is a heterogeneous disease in which immune classifications and four consensus molecular subgroups (CMS) have been described. Our aim was to integrate the composition of the tumor microenvironment with the consensus molecular classification of colorectal cancer. EXPERIMENTAL DESIGN: We retrospectively analyzed the composition and the functional orientation of the immune, fibroblastic, and angiogenic microenvironment of 1,388 colorectal cancer tumors from three independent cohorts using transcriptomics. We validated our findings using immunohistochemistry. RESULTS: We report that colorectal cancer molecular subgroups and microenvironmental signatures are highly correlated. Out of the four molecular subgroups, two highly express immune-specific genes. The good-prognosis microsatellite instable-enriched subgroup (CMS1) is characterized by overexpression of genes specific to cytotoxic lymphocytes. In contrast, the poor-prognosis mesenchymal subgroup (CMS4) expresses markers of lymphocytes and of cells of monocytic origin. The mesenchymal subgroup also displays an angiogenic, inflammatory, and immunosuppressive signature, a coordinated pattern that we also found in breast (n = 254), ovarian (n = 97), lung (n = 80), and kidney (n = 143) cancers. Pathologic examination revealed that the mesenchymal subtype is characterized by a high density of fibroblasts that likely produce the chemokines and cytokines that favor tumor-associated inflammation and support angiogenesis, resulting in a poor prognosis. In contrast, the canonical (CMS2) and metabolic (CMS3) subtypes with intermediate prognosis exhibit low immune and inflammatory signatures. CONCLUSIONS: The distinct immune orientations of the colorectal cancer molecular subtypes pave the way for tailored immunotherapies. Clin Cancer Res; 22(16); 4057-66. ©2016 AACR.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Biomarcadores de Tumor , Fibroblastos Asociados al Cáncer/inmunología , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Análisis por Conglomerados , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/terapia , Perfilación de la Expresión Génica , Humanos , Inmunoterapia , Neovascularización Patológica , Medicina de Precisión , Células del Estroma/inmunología , Células del Estroma/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
14.
Genome Biol ; 17(1): 218, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27765066

RESUMEN

We introduce the Microenvironment Cell Populations-counter (MCP-counter) method, which allows the robust quantification of the absolute abundance of eight immune and two stromal cell populations in heterogeneous tissues from transcriptomic data. We present in vitro mRNA mixture and ex vivo immunohistochemical data that quantitatively support the validity of our method's estimates. Additionally, we demonstrate that MCP-counter overcomes several limitations or weaknesses of previously proposed computational approaches. MCP-counter is applied to draw a global picture of immune infiltrates across human healthy tissues and non-hematopoietic human tumors and recapitulates microenvironment-based patient stratifications associated with overall survival in lung adenocarcinoma and colorectal and breast cancer.


Asunto(s)
Rastreo Celular/métodos , Biología Computacional , Células del Estroma/patología , Microambiente Tumoral/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Biomarcadores de Tumor , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , ARN Mensajero/biosíntesis , Células del Estroma/inmunología , Transcriptoma/genética , Microambiente Tumoral/inmunología
15.
Clin Cancer Res ; 21(13): 3031-40, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25688160

RESUMEN

PURPOSE: Clear cell renal cell carcinoma (ccRCC) has shown durable responses to checkpoint blockade therapies. However, important gaps persist in the understanding of its immune microenvironment. This study aims to investigate the expression and prognostic significance of immune checkpoints in primary and metastatic ccRCC, in relation with mature dendritic cells (DC) and T-cell densities. EXPERIMENTAL DESIGN: We investigated the infiltration and the localization of CD8(+) T cells and mature DC, and the expression of immune checkpoints (PD-1, LAG-3, PD-L1, and PD-L2) in relation with prognosis, in 135 primary ccRCC tumors and 51 ccRCC lung metastases. RNA expression data for 496 primary ccRCC samples were used as confirmatory cohort. RESULTS: We identify two groups of tumors with extensive CD8(+) T-cell infiltrates. One group, characterized by high expression of immune checkpoints in the absence of fully functional mature DC, is associated with increased risk of disease progression. The second group, characterized by low expression of immune checkpoints and localization of mature DC in peritumoral immune aggregates (tertiary lymphoid structures), is associated with good prognosis. CONCLUSIONS: The expression of the immune checkpoints and the localization of DC in the tumor microenvironment modulate the clinical impact of CD8(+) T cells in ccRCC.


Asunto(s)
Carcinoma de Células Renales/inmunología , Neoplasias Renales/inmunología , Antígenos CD/metabolismo , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/fisiología , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/secundario , Supervivencia sin Enfermedad , Humanos , Estimación de Kaplan-Meier , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Linfocitos Infiltrantes de Tumor/fisiología , Análisis Multivariante , Pronóstico , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Modelos de Riesgos Proporcionales , Microambiente Tumoral , Proteína del Gen 3 de Activación de Linfocitos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA