Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pathol ; 245(3): 283-296, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29604063

RESUMEN

Genomic instability, which is a hallmark of cancer, is generally thought to occur in the middle to late stages of tumourigenesis, following the acquisition of permissive molecular aberrations such as TP53 mutation or whole genome doubling. Tumours with somatic POLE exonuclease domain mutations are notable for their extreme genomic instability (their mutation burden is among the highest in human cancer), distinct mutational signature, lymphocytic infiltrate, and excellent prognosis. To what extent these characteristics are determined by the timing of POLE mutations in oncogenesis is unknown. Here, we have shown that pathogenic POLE mutations are detectable in non-malignant precursors of endometrial and colorectal cancer. Using genome and exome sequencing, we found that multiple driver mutations in POLE-mutant cancers show the characteristic POLE mutational signature, including those in genes conventionally regarded as initiators of tumourigenesis. In POLE-mutant cancers, the proportion of monoclonal predicted neoantigens was similar to that in other cancers, but the absolute number was much greater. We also found that the prominent CD8+ T-cell infiltrate present in POLE-mutant cancers was evident in their precursor lesions. Collectively, these data indicate that somatic POLE mutations are early, quite possibly initiating, events in the endometrial and colorectal cancers in which they occur. The resulting early onset of genomic instability may account for the striking immune response and excellent prognosis of these tumours, as well as their early presentation. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Adenocarcinoma/genética , Antígenos de Neoplasias/genética , Neoplasias Colorrectales/genética , ADN Polimerasa II/genética , Neoplasias Endometriales/genética , Mutación , Proteínas de Unión a Poli-ADP-Ribosa/genética , Adenocarcinoma/enzimología , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , ADN Polimerasa II/metabolismo , Bases de Datos Genéticas , Neoplasias Endometriales/enzimología , Neoplasias Endometriales/inmunología , Neoplasias Endometriales/patología , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Inestabilidad Genómica , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Fenotipo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Microambiente Tumoral , Secuenciación Completa del Genoma
2.
J Am Soc Nephrol ; 29(5): 1566-1576, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29610404

RESUMEN

Background Ischemia during kidney transplant causes chronic allograft injury and adversely affects outcome, but the underlying mechanisms are incompletely understood. In tumors, oxygen shortage reduces the DNA demethylating activity of the ten-11 translocation (TET) enzymes, yielding hypermethylated genomes that promote tumor progression. We investigated whether ischemia similarly induces DNA hypermethylation in kidney transplants and contributes to chronic injury.Methods We profiled genome-wide DNA methylation in three cohorts of brain-dead donor kidney allograft biopsy specimens: a longitudinal cohort with paired biopsy specimens obtained at allograft procurement (preischemia; n=13), after implantation and reperfusion (postischemia; n=13), and at 3 or 12 months after transplant (n=5 each); a cross-sectional cohort with preimplantation biopsy specimens (n=82); and a cross-sectional cohort with postreperfusion biopsy specimens (n=46).Results Analysis of the paired preischemia and postischemia specimens revealed that methylation increased drastically in all allografts on ischemia. Hypermethylation was caused by loss of 5-hydroxymethylcytosine, the product of TET activity, and it was stable 1 year after transplant. In the preimplantation cohort, CpG hypermethylation directly correlated with ischemia time and for some CpGs, increased 2.6% per additional hour of ischemia. Hypermethylation preferentially affected and reduced the expression of genes involved in suppressing kidney injury and fibrosis. Moreover, CpG hypermethylation in preimplantation specimens predicted chronic injury, particularly fibrosis and glomerulosclerosis, 1 year after transplant. This finding was validated in the independent postreperfusion cohort, in which hypermethylation also predicted reduced allograft function 1 year after transplant, outperforming established clinical variables.Conclusions We highlight a novel epigenetic basis for ischemia-induced chronic allograft injury with biomarker potential.


Asunto(s)
Aloinjertos/patología , Aloinjertos/fisiopatología , Isquemia Fría/efectos adversos , Metilación de ADN , Isquemia/genética , Isquemia/metabolismo , Trasplante de Riñón , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Adulto , Anciano , Aloinjertos/enzimología , Islas de CpG , Estudios Transversales , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo , Femenino , Fibrosis , Expresión Génica , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/genética , Humanos , Isquemia/complicaciones , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Oxigenasas de Función Mixta/metabolismo , Periodo Posoperatorio , Valor Predictivo de las Pruebas , Periodo Preoperatorio , Proteínas Proto-Oncogénicas/metabolismo
3.
Int J Cancer ; 142(6): 1230-1243, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29063609

RESUMEN

Uterine leiomyosarcomas (uLMS) are rare, aggressive malignancies for which limited treatment options are available. To gain novel molecular insights into uLMS and identify potential novel therapeutic targets, we characterized 84 uLMS samples for genome-wide somatic copy number alterations, mutations, gene fusions and gene expression and performed a data integration analysis. We found that alterations affecting TP53, RB1, PTEN, MED12, YWHAE and VIPR2 were present in the majority of uLMS. Pathway analyses additionally revealed that the PI3K/AKT/mTOR, estrogen-mediated S-phase entry and DNA damage response signaling pathways, for which inhibitors have already been developed and approved, frequently harbored genetic changes. Furthermore, a significant proportion of uLMS was characterized by amplifications and overexpression of known oncogenes (CCNE1, TDO2), as well as deletions and reduced expression of tumor suppressor genes (PTEN, PRDM16). Overall, it emerged that the most frequently affected gene in our uLMS samples was VIPR2 (96%). Interestingly, VIPR2 deletion also correlated with unfavorable survival in uLMS patients (multivariate analysis; HR = 4.5, CI = 1.4-14.3, p = 1.2E-02), while VIPR2 protein expression was reduced in uLMS vs. normal myometrium. Moreover, stimulation of VIPR2 with its natural agonist VIP decreased SK-UT-1 uLMS cell proliferation in a dose-dependent manner. These data suggest that VIPR2, which is a negative regulator of smooth muscle cell proliferation, might be a novel tumor suppressor gene in uLMS. Our work further highlights the importance of integrative molecular analyses, through which we were able to uncover the genes and pathways most frequently affected by somatic alterations in uLMS.


Asunto(s)
Carcinogénesis/genética , Leiomiosarcoma/genética , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Transducción de Señal/genética , Neoplasias Uterinas/genética , Adulto , Anciano , Anciano de 80 o más Años , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Genes Supresores de Tumor , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Estimación de Kaplan-Meier , Leiomiosarcoma/mortalidad , Leiomiosarcoma/patología , Leiomiosarcoma/terapia , Persona de Mediana Edad , Miometrio/patología , Oncogenes/genética , Análisis de Secuencia de ARN/métodos , Neoplasias Uterinas/mortalidad , Neoplasias Uterinas/patología , Neoplasias Uterinas/terapia , Secuenciación Completa del Genoma/métodos
4.
Gynecol Oncol ; 146(3): 538-545, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28625393

RESUMEN

OBJECTIVE: Uterine sarcomas (US) and carcinosarcomas (CS) are rare, aggressive cancers. The lack of reliable preclinical models hampers the search for new treatment strategies and predictive biomarkers. To this end, we established and characterized US and CS patient-derived xenograft (PDX) models. METHODS: Tumor fragments of US and CS were subcutaneously implanted into immunocompromised mice. Engrafted xenograft and original tumors were compared by means of histology, immunohistochemistry, whole-genome low-coverage sequencing for copy number variations, and RNA sequencing. RESULTS: Of 13 implanted leiomyosarcomas (LMS), 10 engrafted (engraftment rate of 77%). Also 2 out of 7 CS (29%) and one high-grade US (not otherwise specified) models were successfully established. LMS xenografts showed high histological similarity to their corresponding human tumors. Expression of desmin and/or H-caldesmon was detected in 8/10 LMS PDX models. We noticed that in CS models, characterized by the concomitant presence of a mesenchymal and an epithelial component, the relative distribution of the components is varying over the generations, as confirmed by changes in vimentin and cytokeratin expression. The similarity in copy number profiles between original and xenograft tumors ranged from 57.7% to 98.2% for LMS models and from 47.4 to 65.8% for CS models. Expression pattern stability was assessed by clustering RNA expression levels of original and xenograft tumors. Six xenografts clustered together with their original tumor, while 3 (all LMS) clustered apart. CONCLUSIONS: We present here a panel of clinically annotated uterine sarcoma and carcinosarcoma PDX models, which will be a useful tool for preclinical testing of new therapies.


Asunto(s)
Carcinosarcoma/patología , ADN de Neoplasias/análisis , Modelos Animales de Enfermedad , Xenoinjertos/patología , Leiomiosarcoma/patología , ARN Neoplásico/análisis , Neoplasias Uterinas/patología , Adulto , Anciano , Animales , Proteínas de Unión a Calmodulina/análisis , Carcinosarcoma/química , Carcinosarcoma/genética , Variaciones en el Número de Copia de ADN , Desmina/análisis , Femenino , Expresión Génica , Supervivencia de Injerto , Xenoinjertos/química , Humanos , Leiomiosarcoma/química , Leiomiosarcoma/genética , Ratones , Persona de Mediana Edad , Trasplante de Neoplasias , Análisis de Secuencia de ARN , Trasplante Heterólogo , Neoplasias Uterinas/química , Neoplasias Uterinas/genética
5.
Endoscopy ; 48(11): 1016-1022, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27626319

RESUMEN

Background and study aim: Typically, pancreatic patient-derived tumor xenografts (PDXs) are established by transplanting large tumor biopsies obtained through invasive surgery approaches into immunocompromised mice. We aimed to develop pancreatic PDXs by transplanting tumor tissue acquired by endoscopic ultrasound (EUS)-guided fine needle biopsies (FNB), assess take rates compared to surgery-derived PDXs, and demonstrate the histological and genetic resemblance to the original tumor. Patients and methods: Biopsies of untreated pancreatic carcinoma were collected at surgery and during EUS and processed to generate PDXs. Results: By centrifugation of FNB-derived tissue prior to engraftment, we achieved an engraftment rate of 60 % (6/10). Despite a decrease in stromal tissue, the general morphology of FNB-derived PDXs was conserved as assessed by histopathology. At the genetic level, somatic mutation and copy number profiles were largely similar to the primary tumor. Conclusion: We show that it is technically feasible to establish pancreatic PDXs using a minimally invasive sampling technique, such as EUS-FNB. Although only a limited amount of tumor tissue was acquired, we obtained results similar to those from surgery-derived PDXs.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico , Trasplante de Neoplasias/métodos , Trasplante de Neoplasias/patología , Neoplasias Pancreáticas/patología , Anciano , Anciano de 80 o más Años , Animales , Carcinoma Ductal Pancreático/genética , Análisis Mutacional de ADN , Exoma , Femenino , Dosificación de Gen , Supervivencia de Injerto , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Mutación , Neoplasias Pancreáticas/genética , Proyectos Piloto , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína p53 Supresora de Tumor/genética
6.
Gynecol Oncol ; 138(1): 165-73, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25933683

RESUMEN

OBJECTIVES: Endometrial carcinoma (EC) is the most common gynecological cancer in the Western World. Treatment options are limited for advanced and recurrent disease. Therefore, new treatment options are necessary. Inhibition of the PI3K/AKT/mTOR and/or the Ras/Raf/MEK pathways is suggested to be clinically relevant. However, the knowledge about the effect of combination targeted therapy in EC is limited. The aim of this study was to investigate the effect of these therapies on primary endometrioid EC cell cultures in vitro and in vivo. METHODS: Primary endometrioid EC cell cultures were incubated with Temsirolimus (mTORC1 inhibitor), NVP-BKM120 (pan-PI3K inhibitor), NVP-BEZ235 (pan-PI3K/mTOR inhibitor), or AZD6244 (MEK1/2 inhibitor) as single treatment. In vitro, the effect of NVP-BEZ235 with or without AZD6244 was determined for cell viability, cell cycle arrest, apoptosis induction, and cell signaling. In vivo, the effect of NVP-BEZ35 was investigated for 2 subcutaneous xenograft models of the corresponding primary cultures. RESULTS: NVP-BEZ235 was the most potent PI3K/AKT/mTOR pathway inhibitor. NVP-BEZ235 and AZD6244 reduced cell viability and induced cell cycle arrest and apoptosis, by reduction of p-AKT, p-S6, and p-ERK levels. Combination treatment showed a synergistic effect. In vivo, NVP-BEZ235 reduced tumor growth and inhibited p-S6 expression. The effects of the compounds were independent of the mutation profile of the cell cultures used. CONCLUSIONS: A synergistic antitumor effect was shown for NVP-BEZ235 and AZD6244 in primary endometrioid EC cells in vitro. In addition, NVP-BEZ235 induced reduction of tumor growth in vivo. Therefore, targeted therapies seem an interesting strategy to further evaluate in clinical trials.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bencimidazoles/farmacología , Carcinoma Endometrioide/tratamiento farmacológico , Neoplasias Endometriales/tratamiento farmacológico , Imidazoles/farmacología , Quinolinas/farmacología , Animales , Bencimidazoles/administración & dosificación , Carcinoma Endometrioide/enzimología , Sinergismo Farmacológico , Neoplasias Endometriales/enzimología , Femenino , Humanos , Imidazoles/administración & dosificación , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinolinas/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas raf/antagonistas & inhibidores , Quinasas raf/metabolismo , Proteínas ras/antagonistas & inhibidores , Proteínas ras/metabolismo
7.
Gynecol Oncol ; 139(1): 118-26, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26232337

RESUMEN

OBJECTIVE: Endometrial carcinoma (EC) is the sixth most common cancer in women and therapies are limited for advanced and recurrent disease. Patient-derived tumor xenograft (PDTX) models are becoming popular tools in translational research because of their histological and genetic similarity to the original tumors and the ability to predict therapeutic response to treatments. Here, we established and characterized a panel of 24 EC PDTX models which includes the major histological and genetic subtypes observed in patients. METHODS: Fresh tumor tissues collected from primary, metastatic and recurrent type I and type II EC patients were engrafted in immunocompromised mice. Histology, vimentin, and cytokeratin expression were evaluated, together with Microsatellite instability (MSI), mutation profiling by Whole Exome Sequencing and copy number profiling by Whole Genome Low Coverage Sequencing. The efficacy of both PI3K and MEK inhibitors was evaluated in a model of endometrioid carcinoma harboring PTEN, PIK3CA and KRAS mutations. RESULTS: We observed good similarity between primary tumors and the corresponding xenografts, at histological and genetic level. Among the engrafted endometrioid models, we found a significant enrichment of MSI and POLE mutated tumors, compared to non-engrafted samples. Combination treatment with NVP-BEZ235 and AZD6244 showed the possibility to stabilize the tumor growth in one model originated from a patient who already received several lines of chemotherapy. CONCLUSION: The established EC PDTX models, resembling the original human tumors, promise to be useful for preclinical evaluation of novel combination and targeted therapies in specific EC subgroups.


Asunto(s)
Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Neoplasias Endometriales/enzimología , Neoplasias Endometriales/genética , Femenino , Humanos , Ratones , Terapia Molecular Dirigida , Trasplante de Neoplasias , Inhibidores de Proteínas Quinasas/farmacología
8.
Int J Gynecol Cancer ; 25(3): 363-71, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25695543

RESUMEN

OBJECTIVE: Well-characterized, low-passage, primary cell cultures established directly from patient tumors are an important tool for drug screening because these cultures faithfully recapitulate the genomic features of primary tumors. Here, we aimed to establish these cell cultures from primary endometrial carcinomas (ECs) and to develop subcutaneous and orthotopic xenograft models as a model to validate promising treatment options for EC in the in vivo setting. METHODS: Primary cell cultures of EC tumors were established and validated by analysing histologic and genetic characteristics, telomerase activity, and in vitro and in vivo growth characteristics. Using these primary cell cultures, subcutaneous and orthotopic mouse models were subsequently established. RESULTS: We established and characterized 7 primary EC cell cultures and corresponding xenograft models of different types of endometrioid tumors. Interestingly, we observed that the chance to successfully establish a primary cell culture seems higher for microsatellite instable than microsatellite stable tumors. For the first time, we also established an orthotopic murine model for EC derived from a primary cell culture. In contrast to EC cell lines, grafted tumor cultures preserved the original tumor structure and mimicked all histologic features. They also established abdominal and distant metastases, reflecting the tumorigenic behavior in the clinical setting. Remarkably, the established cell cultures and xenograft tumors also preserved the genetic characteristics of the primary tumor. CONCLUSIONS: The established EC cultures reflect the epithelial genetic characteristics of the primary tumor. Therefore, they provide an appropriate model to investigate EC biology and apply high-throughput drug screening experiments. In addition, the established murine xenograft models, in particular the orthotopic model, will be useful to validate promising therapeutic strategies in vivo, as the grafted tumors closely resemble the primary tumors from which they were derived. Microsatellite instable status seems to determine the success rate of establishing primary cell cultures.


Asunto(s)
Carcinoma/genética , Modelos Animales de Enfermedad , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Inestabilidad de Microsatélites , Adulto , Anciano , Animales , Carcinoma/química , Carcinoma/secundario , Proliferación Celular , Neoplasias Endometriales/química , Femenino , Humanos , Queratinas/análisis , Ratones , Ratones Desnudos , Repeticiones de Microsatélite , Persona de Mediana Edad , Trasplante de Neoplasias/métodos , Cultivo Primario de Células , Receptores de Estrógenos/análisis , Receptores de Progesterona/análisis , Células Tumorales Cultivadas , Vimentina/análisis
9.
Nat Commun ; 9(1): 4112, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30291241

RESUMEN

Increased copy number alterations (CNAs) indicative of chromosomal instability (CIN) have been associated with poor cancer outcome. Here, we study CNAs as potential biomarkers of bevacizumab (BVZ) response in metastatic colorectal cancer (mCRC). We cluster 409 mCRCs in three subclusters characterized by different degrees of CIN. Tumors belonging to intermediate-to-high instability clusters have improved outcome following chemotherapy plus BVZ versus chemotherapy alone. In contrast, low instability tumors, which amongst others consist of POLE-mutated and microsatellite-instable tumors, derive no further benefit from BVZ. This is confirmed in 81 mCRC tumors from the phase 2 MoMa study involving BVZ. CNA clusters overlap with CRC consensus molecular subtypes (CMS); CMS2/4 xenografts correspond to intermediate-to-high instability clusters and respond to FOLFOX chemotherapy plus mouse avastin (B20), while CMS1/3 xenografts match with low instability clusters and fail to respond. Overall, we identify copy number load as a novel potential predictive biomarker of BVZ combination therapy.


Asunto(s)
Adenocarcinoma/genética , Antineoplásicos Inmunológicos/uso terapéutico , Bevacizumab/uso terapéutico , Neoplasias Colorrectales/genética , Variaciones en el Número de Copia de ADN , Adenocarcinoma/tratamiento farmacológico , Anciano , Animales , Inestabilidad Cromosómica , Neoplasias Colorrectales/tratamiento farmacológico , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Estudios Retrospectivos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Nat Commun ; 9(1): 3166, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30093612

RESUMEN

Endometrial cancer is the most commonly diagnosed cancer of the female reproductive tract in developed countries. Through genome-wide association studies (GWAS), we have previously identified eight risk loci for endometrial cancer. Here, we present an expanded meta-analysis of 12,906 endometrial cancer cases and 108,979 controls (including new genotype data for 5624 cases) and identify nine novel genome-wide significant loci, including a locus on 12q24.12 previously identified by meta-GWAS of endometrial and colorectal cancer. At five loci, expression quantitative trait locus (eQTL) analyses identify candidate causal genes; risk alleles at two of these loci associate with decreased expression of genes, which encode negative regulators of oncogenic signal transduction proteins (SH2B3 (12q24.12) and NF1 (17q11.2)). In summary, this study has doubled the number of known endometrial cancer risk loci and revealed candidate causal genes for future study.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Endometriales/genética , Predisposición Genética a la Enfermedad , Alelos , Cromatina/química , Femenino , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Factores de Riesgo , Transducción de Señal
11.
Cancer Med ; 7(5): 1978-1987, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29608257

RESUMEN

Epidemiological, biological, and molecular data suggest links between endometriosis and endometrial cancer, with recent epidemiological studies providing evidence for an association between a previous diagnosis of endometriosis and risk of endometrial cancer. We used genetic data as an alternative approach to investigate shared biological etiology of these two diseases. Genetic correlation analysis of summary level statistics from genomewide association studies (GWAS) using LD Score regression revealed moderate but significant genetic correlation (rg  = 0.23, P = 9.3 × 10-3 ), and SNP effect concordance analysis provided evidence for significant SNP pleiotropy (P = 6.0 × 10-3 ) and concordance in effect direction (P = 2.0 × 10-3 ) between the two diseases. Cross-disease GWAS meta-analysis highlighted 13 distinct loci associated at P ≤ 10-5 with both endometriosis and endometrial cancer, with one locus (SNP rs2475335) located within PTPRD associated at a genomewide significant level (P = 4.9 × 10-8 , OR = 1.11, 95% CI = 1.07-1.15). PTPRD acts in the STAT3 pathway, which has been implicated in both endometriosis and endometrial cancer. This study demonstrates the value of cross-disease genetic analysis to support epidemiological observations and to identify biological pathways of relevance to multiple diseases.


Asunto(s)
Neoplasias Endometriales/genética , Endometriosis/genética , Predisposición Genética a la Enfermedad/genética , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Australia/epidemiología , Neoplasias Endometriales/epidemiología , Endometriosis/epidemiología , Endometrio/patología , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Polimorfismo de Nucleótido Simple/genética , Factor de Transcripción STAT3/metabolismo
12.
Clin Cancer Res ; 23(23): 7232-7241, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28939739

RESUMEN

Purpose: Molecular classification of endometrial cancer identified distinct molecular subgroups. However, the largest subset of endometrial cancers remains poorly characterized and is referred to as the "nonspecific molecular profile" (NSMP) subgroup. Here, we aimed at refining the classification of this subgroup by profiling somatic copy-number aberrations (SCNAs).Experimental Design: SCNAs were analyzed in 141 endometrial cancers using whole-genome SNP arrays and pooled with 361 endometrial cancers from The Cancer Genome Atlas. Genomic Identification of Significant Targets in Cancer (GISTIC) identified statistically enriched SCNAs and penalized Cox regression assessed survival effects. The prognostic significance of relevant SCNAs was validated using multiplex ligation-dependent probe amplification in 840 endometrial cancers from the PORTEC-1/2 trials. Copy-number status of genes was correlated with gene expression to identify potential cancer drivers. One plausible oncogene was validated in vitro using antisense oligonucleotide-based strategy.Results: SCNAs affecting chromosome 1q32.1 significantly correlated with worse relapse-free survival (RFS) in the NSMP subgroup (HR, 2.12; 95% CI, 1.26-3.59; P = 0.005). This effect was replicated in NSMP endometrial cancers from PORTEC-1/2 (HR, 2.34; 95% CI, 1.17-4.70; P = 0.017). A new molecular classification including the 1q32.1 amplification improved risk prediction of recurrence. MDM4 gene expression strongly correlated with 1q32.1 amplification. Silencing MDM4 inhibited cell growth in cell lines carrying 1q32.1 amplification, but not in those without MDM4 amplification. Vice versa, increasing MDM4 expression in nonamplified cell lines stimulated cell proliferation.Conclusions: 1q32.1 amplification was identified as a prognostic marker for poorly characterized NSMP endometrial cancers, refining the molecular classification of this subgroup. We functionally validated MDM4 as a potential oncogenic driver in the 1q32.1 region. Clin Cancer Res; 23(23); 7232-41. ©2017 AACR.


Asunto(s)
Cromosomas Humanos Par 1/genética , Variaciones en el Número de Copia de ADN/genética , Neoplasias Endometriales/genética , Amplificación de Genes , Anciano , Línea Celular Tumoral , Análisis por Conglomerados , Supervivencia sin Enfermedad , Neoplasias Endometriales/patología , Neoplasias Endometriales/terapia , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Evaluación de Resultado en la Atención de Salud/métodos , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Pronóstico , Modelos de Riesgos Proporcionales
13.
Anticancer Res ; 36(10): 5381-5384, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27798902

RESUMEN

BACKGROUND/AIM: Human epidermal growth factor receptor 2 (HER2) positivity is associated with a worse prognosis in endometrial cancer (EC). Trastuzumab as a single agent did not demonstrate activity in such cases but there are no reports on its combined use with taxanes. We report the outcome in patients treated simultaneously with trastuzumab and paclitaxel for advanced or recurrent HER2-positive endometrial carcinoma and compared it to their microsatellite instability (MSI) status and PIK3CA mutational profiles. PATIENTS AND METHODS: Patients with advancedor recurrent endometrial carcinoma showing HER2 overexpression (2+ or 3+ immunohistochemical staining) or HER2 amplification (fluorescence in situ hybridization (FISH) HER2/chromosome 17 centromere (CEP 17) ratio >2.0) were treated with trastuzumab (8 mg/kg) and paclitaxel (90 mg/m2) every three weeks. Evaluation of the response was assessed according to the response evaluation criteria in solid tumors (RECIST) guidelines. Endometrial tumors, sampled before the beginning of trastuzumab, were genotyped for PIK3CA hot spot mutations using Sequenom iPLEX Assay technology. RESULTS: Two uterine serous adenocarcinomas and one grade 3 endometrioid adenocarcinoma showing HER2 positivity were treated with trastuzumab and paclitaxel. Between three and seven months of treatment, the three cases showed progressive disease. The genomic analysis of the three cases showed different mutational profiles. One case was found to have MSI and had one PIK3CA mutation. The two others showed no hot spot mutation for PIK3CA. CONCLUSION: Even associated with paclitaxel, HER2-positive endometrial carcinomas poorly responded to trastuzumab. This report underlines the low accuracy of HER2 positivity to predict response of endometrial cancer to combined targeted therapy using trastuzumab and paclitaxel.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Endometriales/tratamiento farmacológico , Genes erbB-2 , Anciano , Neoplasias Endometriales/genética , Femenino , Humanos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Paclitaxel/administración & dosificación , Trastuzumab/administración & dosificación
14.
Endocr Relat Cancer ; 23(2): 77-91, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26574572

RESUMEN

Candidate gene studies have reported CYP19A1 variants to be associated with endometrial cancer and with estradiol (E2) concentrations. We analyzed 2937 single nucleotide polymorphisms (SNPs) in 6608 endometrial cancer cases and 37 925 controls and report the first genome wide-significant association between endometrial cancer and a CYP19A1 SNP (rs727479 in intron 2, P=4.8×10(-11)). SNP rs727479 was also among those most strongly associated with circulating E2 concentrations in 2767 post-menopausal controls (P=7.4×10(-8)). The observed endometrial cancer odds ratio per rs727479 A-allele (1.15, CI=1.11-1.21) is compatible with that predicted by the observed effect on E2 concentrations (1.09, CI=1.03-1.21), consistent with the hypothesis that endometrial cancer risk is driven by E2. From 28 candidate-causal SNPs, 12 co-located with three putative gene-regulatory elements and their risk alleles associated with higher CYP19A1 expression in bioinformatical analyses. For both phenotypes, the associations with rs727479 were stronger among women with a higher BMI (Pinteraction=0.034 and 0.066 respectively), suggesting a biologically plausible gene-environment interaction.


Asunto(s)
Aromatasa/genética , Neoplasias Endometriales/etiología , Estradiol/sangre , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Genotipo , Polimorfismo de Nucleótido Simple , Factores de Edad , Alelos , Índice de Masa Corporal , Estudios de Casos y Controles , Neoplasias Endometriales/sangre , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Femenino , Estudios de Asociación Genética , Humanos , Fenotipo
15.
Nat Genet ; 48(6): 667-674, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27135401

RESUMEN

We conducted a meta-analysis of three endometrial cancer genome-wide association studies (GWAS) and two follow-up phases totaling 7,737 endometrial cancer cases and 37,144 controls of European ancestry. Genome-wide imputation and meta-analysis identified five new risk loci of genome-wide significance at likely regulatory regions on chromosomes 13q22.1 (rs11841589, near KLF5), 6q22.31 (rs13328298, in LOC643623 and near HEY2 and NCOA7), 8q24.21 (rs4733613, telomeric to MYC), 15q15.1 (rs937213, in EIF2AK4, near BMF) and 14q32.33 (rs2498796, in AKT1, near SIVA1). We also found a second independent 8q24.21 signal (rs17232730). Functional studies of the 13q22.1 locus showed that rs9600103 (pairwise r(2) = 0.98 with rs11841589) is located in a region of active chromatin that interacts with the KLF5 promoter region. The rs9600103[T] allele that is protective in endometrial cancer suppressed gene expression in vitro, suggesting that regulation of the expression of KLF5, a gene linked to uterine development, is implicated in tumorigenesis. These findings provide enhanced insight into the genetic and biological basis of endometrial cancer.


Asunto(s)
Neoplasias Endometriales/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Cromosomas Humanos Par 8 , Femenino , Humanos , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas
16.
Cancer Epidemiol Biomarkers Prev ; 25(11): 1503-1510, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27550749

RESUMEN

BACKGROUND: The strongest known risk factor for endometrial cancer is obesity. To determine whether SNPs associated with increased body mass index (BMI) or waist-hip ratio (WHR) are associated with endometrial cancer risk, independent of measured BMI, we investigated relationships between 77 BMI and 47 WHR SNPs and endometrial cancer in 6,609 cases and 37,926 country-matched controls. METHODS: Logistic regression analysis and fixed effects meta-analysis were used to test for associations between endometrial cancer risk and (i) individual BMI or WHR SNPs, (ii) a combined weighted genetic risk score (wGRS) for BMI or WHR. Causality of BMI for endometrial cancer was assessed using Mendelian randomization, with BMIwGRS as instrumental variable. RESULTS: The BMIwGRS was significantly associated with endometrial cancer risk (P = 3.4 × 10-17). Scaling the effect of the BMIwGRS on endometrial cancer risk by its effect on BMI, the endometrial cancer OR per 5 kg/m2 of genetically predicted BMI was 2.06 [95% confidence interval (CI), 1.89-2.21], larger than the observed effect of BMI on endometrial cancer risk (OR = 1.55; 95% CI, 1.44-1.68, per 5 kg/m2). The association attenuated but remained significant after adjusting for BMI (OR = 1.22; 95% CI, 1.10-1.39; P = 5.3 × 10-4). There was evidence of directional pleiotropy (P = 1.5 × 10-4). BMI SNP rs2075650 was associated with endometrial cancer at study-wide significance (P < 4.0 × 10-4), independent of BMI. Endometrial cancer was not significantly associated with individual WHR SNPs or the WHRwGRS. CONCLUSIONS: BMI, but not WHR, is causally associated with endometrial cancer risk, with evidence that some BMI-associated SNPs alter endometrial cancer risk via mechanisms other than measurable BMI. IMPACT: The causal association between BMI SNPs and endometrial cancer has possible implications for endometrial cancer risk modeling. Cancer Epidemiol Biomarkers Prev; 25(11); 1503-10. ©2016 AACR.


Asunto(s)
Índice de Masa Corporal , Neoplasias Endometriales/etiología , Predisposición Genética a la Enfermedad , Análisis de la Aleatorización Mendeliana , Obesidad/complicaciones , Polimorfismo de Nucleótido Simple , Neoplasias Endometriales/genética , Femenino , Humanos , Obesidad/genética , Relación Cintura-Cadera
17.
J Natl Cancer Inst ; 107(1): 402, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25505230

RESUMEN

BACKGROUND: Current risk stratification in endometrial cancer (EC) results in frequent over- and underuse of adjuvant therapy, and may be improved by novel biomarkers. We examined whether POLE proofreading mutations, recently reported in about 7% of ECs, predict prognosis. METHODS: We performed targeted POLE sequencing in ECs from the PORTEC-1 and -2 trials (n = 788), and analyzed clinical outcome according to POLE status. We combined these results with those from three additional series (n = 628) by meta-analysis to generate multivariable-adjusted, pooled hazard ratios (HRs) for recurrence-free survival (RFS) and cancer-specific survival (CSS) of POLE-mutant ECs. All statistical tests were two-sided. RESULTS: POLE mutations were detected in 48 of 788 (6.1%) ECs from PORTEC-1 and-2 and were associated with high tumor grade (P < .001). Women with POLE-mutant ECs had fewer recurrences (6.2% vs 14.1%) and EC deaths (2.3% vs 9.7%), though, in the total PORTEC cohort, differences in RFS and CSS were not statistically significant (multivariable-adjusted HR = 0.43, 95% CI = 0.13 to 1.37, P = .15; HR = 0.19, 95% CI = 0.03 to 1.44, P = .11 respectively). However, of 109 grade 3 tumors, 0 of 15 POLE-mutant ECs recurred, compared with 29 of 94 (30.9%) POLE wild-type cancers; reflected in statistically significantly greater RFS (multivariable-adjusted HR = 0.11, 95% CI = 0.001 to 0.84, P = .03). In the additional series, there were no EC-related events in any of 33 POLE-mutant ECs, resulting in a multivariable-adjusted, pooled HR of 0.33 for RFS (95% CI = 0.12 to 0.91, P = .03) and 0.26 for CSS (95% CI = 0.06 to 1.08, P = .06). CONCLUSION: POLE proofreading mutations predict favorable EC prognosis, independently of other clinicopathological variables, with the greatest effect seen in high-grade tumors. This novel biomarker may help to reduce overtreatment in EC.


Asunto(s)
Biomarcadores de Tumor/genética , ADN Polimerasa II/genética , Neoplasias Endometriales/genética , Neoplasias Endometriales/mortalidad , Mutación , Adulto , Supervivencia sin Enfermedad , Neoplasias Endometriales/patología , Femenino , Humanos , Estimación de Kaplan-Meier , Clasificación del Tumor , Oportunidad Relativa , Proteínas de Unión a Poli-ADP-Ribosa , Valor Predictivo de las Pruebas , Pronóstico
18.
Endocr Relat Cancer ; 22(5): 851-61, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26330482

RESUMEN

Excessive exposure to estrogen is a well-established risk factor for endometrial cancer (EC), particularly for cancers of endometrioid histology. The physiological function of estrogen is primarily mediated by estrogen receptor alpha, encoded by ESR1. Consequently, several studies have investigated whether variation at the ESR1 locus is associated with risk of EC, with conflicting results. We performed comprehensive fine-mapping analyses of 3633 genotyped and imputed single nucleotide polymorphisms (SNPs) in 6607 EC cases and 37 925 controls. There was evidence of an EC risk signal located at a potential alternative promoter of the ESR1 gene (lead SNP rs79575945, P=1.86×10(-5)), which was stronger for cancers of endometrioid subtype (P=3.76×10(-6)). Bioinformatic analysis suggests that this risk signal is in a functionally important region targeting ESR1, and eQTL analysis found that rs79575945 was associated with expression of SYNE1, a neighbouring gene. In summary, we have identified a single EC risk signal located at ESR1, at study-wide significance. Given SNPs located at this locus have been associated with risk for breast cancer, also a hormonally driven cancer, this study adds weight to the rationale for performing informed candidate fine-scale genetic studies across cancer types.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Endometriales/genética , Receptor alfa de Estrógeno/genética , Predisposición Genética a la Enfermedad , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple/genética , Estudios de Casos y Controles , Biología Computacional , Proteínas del Citoesqueleto , Bases de Datos Genéticas , Femenino , Sitios Genéticos , Genotipo , Humanos , Metaanálisis como Asunto , Pronóstico , Regiones Promotoras Genéticas/genética , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA