Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pathol ; 263(2): 217-225, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38551073

RESUMEN

Environmental factors like the pathogenicity island polyketide synthase positive (pks+) Escherichia coli (E. coli) could have potential for risk stratification in colorectal cancer (CRC) screening. The association between pks+ E. coli measured in fecal immunochemical test (FIT) samples and the detection of advanced neoplasia (AN) at colonoscopy was investigated. Biobanked FIT samples were analyzed for both presence of E. coli and pks+ E. coli and correlated with colonoscopy findings; 5020 CRC screening participants were included. Controls were participants in which no relevant lesion was detected because of FIT-negative results (cut-off ≥15 µg Hb/g feces), a negative colonoscopy, or a colonoscopy during which only a nonadvanced polyp was detected. Cases were participants with AN [CRC, advanced adenoma (AA), or advanced serrated polyp (ASP)]. Existing DNA isolation and quantitative polymerase chain reaction (qPCR) procedures were used for the detection of E. coli and pks+ E. coli in stool. A total of 4542 (90.2%) individuals were E. coli positive, and 1322 (26.2%) were pks+ E. coli positive. The prevalence of E. coli in FIT samples from individuals with AN was 92.9% compared to 89.7% in FIT samples of controls (p = 0.010). The prevalence of pks+ E. coli in FIT samples from individuals with AN (28.6%) and controls (25.9%) was not significantly different (p = 0.13). The prevalences of pks+ E. coli in FIT samples from individuals with CRC, AA, or ASP were 29.6%, 28.3%, and 32.1%, respectively. In conclusion, the prevalence of pks+ E. coli in a screening population was 26.2% and did not differ significantly between individuals with AN and controls. These findings disqualify the straightforward option of using a snapshot measurement of pks+ E. coli in FIT samples as a stratification biomarker for CRC risk. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias Colorrectales , Detección Precoz del Cáncer , Escherichia coli , Heces , Sintasas Poliquetidas , Humanos , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/diagnóstico , Heces/microbiología , Heces/enzimología , Escherichia coli/aislamiento & purificación , Escherichia coli/enzimología , Escherichia coli/genética , Masculino , Detección Precoz del Cáncer/métodos , Femenino , Persona de Mediana Edad , Anciano , Sintasas Poliquetidas/genética , Colonoscopía , Factores de Riesgo , Adenoma/microbiología , Adenoma/diagnóstico , Medición de Riesgo , Biomarcadores de Tumor , Estudios de Casos y Controles
2.
Int J Cancer ; 146(7): 1979-1992, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31411736

RESUMEN

Removal of colorectal adenomas is an effective strategy to reduce colorectal cancer (CRC) mortality rates. However, as only a minority of adenomas progress to cancer, such strategies may lead to overtreatment. The present study aimed to characterize adenomas by in-depth molecular profiling, to obtain insights into altered biology associated with the colorectal adenoma-to-carcinoma progression. We obtained low-coverage whole genome sequencing, RNA sequencing and tandem mass spectrometry data for 30 CRCs, 30 adenomas and 18 normal adjacent colon samples. These data were used for DNA copy number aberrations profiling, differential expression, gene set enrichment and gene-dosage effect analysis. Protein expression was independently validated by immunohistochemistry on tissue microarrays and in patient-derived colorectal adenoma organoids. Stroma percentage was determined by digital image analysis of tissue sections. Twenty-four out of 30 adenomas could be unambiguously classified as high risk (n = 9) or low risk (n = 15) of progressing to cancer, based on DNA copy number profiles. Biological processes more prevalent in high-risk than low-risk adenomas were related to proliferation, tumor microenvironment and Notch, Wnt, PI3K/AKT/mTOR and Hedgehog signaling, while metabolic processes and protein secretion were enriched in low-risk adenomas. DNA copy number driven gene-dosage effect in high-risk adenomas and cancers was observed for POFUT1, RPRD1B and EIF6. Increased POFUT1 expression in high-risk adenomas was validated in tissue samples and organoids. High POFUT1 expression was also associated with Notch signaling enrichment and with decreased goblet cells differentiation. In-depth molecular characterization of colorectal adenomas revealed POFUT1 and Notch signaling as potential drivers of tumor progression.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Fucosiltransferasas/genética , Proteínas Oncogénicas/genética , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Biomarcadores de Tumor , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patología , Neoplasias Colorrectales/metabolismo , Progresión de la Enfermedad , Fucosiltransferasas/metabolismo , Humanos , Proteínas Oncogénicas/metabolismo , Reproducibilidad de los Resultados , Microambiente Tumoral
3.
BMC Gastroenterol ; 19(1): 146, 2019 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-31420015

RESUMEN

BACKGROUND: Microvessel density (MVD), as a derived marker for angiogenesis, has been associated with poor outcome in several types of cancer. This study aimed to evaluate the prognostic value of MVD in stage II and III colon cancer and its relation to tumour-stroma-percentage (TSP) and expression of HIF1A and VEGFA. METHODS: Formalin-fixed paraffin-embedded (FFPE) colon cancer tissues were collected from 53 stage II and 54 (5-fluorouracil-treated) stage III patients. MVD was scored by digital morphometric analysis of CD31-stained whole tumour sections. TSP was scored using haematoxylin-eosin stained slides. Protein expression of HIF1A and VEGFA was determined by immunohistochemical evaluation of tissue microarrays. RESULTS: Median MVD was higher in stage III compared to stage II colon cancers (11.1% versus 5.6% CD31-positive tissue area, p < 0.001). High MVD in stage II patients tended to be associated with poor disease free survival (DFS) in univariate analysis (p = 0.056). In contrast, high MVD in 5FU-treated stage III patients was associated with better DFS (p = 0.006). Prognostic value for MVD was observed in multivariate analyses for both cancer stages. CONCLUSIONS: MVD is an independent prognostic factor associated with poor DFS in stage II colon cancer patients, and with better DFS in stage III colon cancer patients treated with adjuvant chemotherapy.


Asunto(s)
Quimioterapia Adyuvante/métodos , Neoplasias del Colon , Microvasos , Neovascularización Patológica , Colon/patología , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/patología , Densitometría/métodos , Femenino , Humanos , Interpretación de Imagen Asistida por Computador/métodos , Inmunohistoquímica , Masculino , Microvasos/diagnóstico por imagen , Microvasos/patología , Persona de Mediana Edad , Estadificación de Neoplasias , Neovascularización Patológica/diagnóstico por imagen , Neovascularización Patológica/etiología , Países Bajos , Pronóstico , Reproducibilidad de los Resultados , Estudios Retrospectivos
4.
J Pathol ; 246(3): 266-276, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29968252

RESUMEN

Consensus molecular subtyping is an RNA expression-based classification system for colorectal cancer (CRC). Genomic alterations accumulate during CRC pathogenesis, including the premalignant adenoma stage, leading to changes in RNA expression. Only a minority of adenomas progress to malignancies, a transition that is associated with specific DNA copy number aberrations or microsatellite instability (MSI). We aimed to investigate whether colorectal adenomas can already be stratified into consensus molecular subtype (CMS) classes, and whether specific CMS classes are related to the presence of specific DNA copy number aberrations associated with progression to malignancy. RNA sequencing was performed on 62 adenomas and 59 CRCs. MSI status was determined with polymerase chain reaction-based methodology. DNA copy number was assessed by low-coverage DNA sequencing (n = 30) or array-comparative genomic hybridisation (n = 32). Adenomas were classified into CMS classes together with CRCs from the study cohort and from The Cancer Genome Atlas (n = 556), by use of the established CMS classifier. As a result, 54 of 62 (87%) adenomas were classified according to the CMS. The CMS3 'metabolic subtype', which was least common among CRCs, was most prevalent among adenomas (n = 45; 73%). One of the two adenomas showing MSI was classified as CMS1 (2%), the 'MSI immune' subtype. Eight adenomas (13%) were classified as the 'canonical' CMS2. No adenomas were classified as the 'mesenchymal' CMS4, consistent with the fact that adenomas lack invasion-associated stroma. The distribution of the CMS classes among adenomas was confirmed in an independent series. CMS3 was enriched with adenomas at low risk of progressing to CRC, whereas relatively more high-risk adenomas were observed in CMS2. We conclude that adenomas can be stratified into the CMS classes. Considering that CMS1 and CMS2 expression signatures may mark adenomas at increased risk of progression, the distribution of the CMS classes among adenomas is consistent with the proportion of adenomas expected to progress to CRC. © 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)
Adenoma/genética , Biomarcadores de Tumor/genética , Carcinoma/genética , Neoplasias Colorrectales/genética , Variaciones en el Número de Copia de ADN , Dosificación de Gen , Perfilación de la Expresión Génica/métodos , Inestabilidad de Microsatélites , Adenoma/clasificación , Adenoma/metabolismo , Carcinoma/clasificación , Carcinoma/metabolismo , Diferenciación Celular , Neoplasias Colorrectales/clasificación , Neoplasias Colorrectales/patología , Consenso , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Estadificación de Neoplasias , Fenotipo , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Transcriptoma
5.
Mol Cell Proteomics ; 16(10): 1850-1863, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28747380

RESUMEN

Proteogenomics, i.e. comprehensive integration of genomics and proteomics data, is a powerful approach identifying novel protein biomarkers. This is especially the case for proteins that differ structurally between disease and control conditions. As tumor development is associated with aberrant splicing, we focus on this rich source of cancer specific biomarkers. To this end, we developed a proteogenomic pipeline, Splicify, which can detect differentially expressed protein isoforms. Splicify is based on integrating RNA massive parallel sequencing data and tandem mass spectrometry proteomics data to identify protein isoforms resulting from differential splicing between two conditions. Proof of concept was obtained by applying Splicify to RNA sequencing and mass spectrometry data obtained from colorectal cancer cell line SW480, before and after siRNA-mediated downmodulation of the splicing factors SF3B1 and SRSF1. These analyses revealed 2172 and 149 differentially expressed isoforms, respectively, with peptide confirmation upon knock-down of SF3B1 and SRSF1 compared with their controls. Splice variants identified included RAC1, OSBPL3, MKI67, and SYK. One additional sample was analyzed by PacBio Iso-Seq full-length transcript sequencing after SF3B1 downmodulation. This analysis verified the alternative splicing identified by Splicify and in addition identified novel splicing events that were not represented in the human reference genome annotation. Therefore, Splicify offers a validated proteogenomic data analysis pipeline for identification of disease specific protein biomarkers resulting from mRNA alternative splicing. Splicify is publicly available on GitHub (https://github.com/NKI-TGO/SPLICIFY) and suitable to address basic research questions using pre-clinical model systems as well as translational research questions using patient-derived samples, e.g. allowing to identify clinically relevant biomarkers.


Asunto(s)
Empalme Alternativo , Biomarcadores de Tumor/análisis , Proteogenómica/métodos , Proteoma/análisis , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Conformación Proteica , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Proteoma/genética , Empalme del ARN , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Análisis de Secuencia de ARN , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo
6.
Br J Cancer ; 115(12): 1565-1574, 2016 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-27855440

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is the third most common cancer worldwide. Accurately identifying stage II CRC patients at risk for recurrence is an unmet clinical need. KCNQ1 was previously identified as a tumour suppressor gene and loss of expression was associated with poor survival in patients with CRC liver metastases. In this study the prognostic value of KCNQ1 in stage II and stage III colon cancer patients was examined. METHODS: KCNQ1 mRNA expression was assessed in 90 stage II colon cancer patients (AMC-AJCCII-90) using microarray gene expression data. Subsequently, KCNQ1 protein expression was evaluated in an independent cohort of 386 stage II and stage III colon cancer patients by immunohistochemistry of tissue microarrays. RESULTS: Low KCNQ1 mRNA expression in stage II microsatellite stable (MSS) colon cancers was associated with poor disease-free survival (DFS) (P=0.025). Loss of KCNQ1 protein expression from epithelial cells was strongly associated with poor DFS in stage II MSS (P<0.0001), stage III MSS (P=0.0001) and stage III microsatellite instable colon cancers (P=0.041). KCNQ1 seemed an independent prognostic value in addition to other high-risk parameters like angio-invasion, nodal stage and microsatellite instability-status. CONCLUSIONS: We conclude that KCNQ1 is a promising biomarker for prediction of disease recurrence and may aid stratification of patients with stage II MSS colon cancer for adjuvant chemotherapy.


Asunto(s)
Neoplasias del Colon/genética , Canal de Potasio KCNQ1/genética , Recurrencia Local de Neoplasia , Neoplasias del Colon/patología , Humanos , Pronóstico , ARN Mensajero/genética
7.
Ann Surg ; 263(1): 138-45, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25563886

RESUMEN

OBJECTIVE: To investigate the individual and combined prognostic value of HIF1α, SLC2A1, and vascular endothelial growth factor A (VEGFA) in a multi-institutional cohort of patients with resected colorectal cancer liver metastasis (CRCLM). BACKGROUND: In the majority of patients with CRCLM, resection seems not to be curative, despite its curative intent. Overexpression of hypoxia-inducible factor 1α (HIF1α), glucose transporter 1 (SLC2A1; also known as GLUT1), and VEGFA has been associated with tumor progression and poor prognosis of patients with colorectal cancer (CRC). METHODS: Tissue microarrays were generated using CRCLM and patient-matched primary CRC from patients who underwent CRCLM resection between 1990 and 2010. Prognostic value of HIF1α, SLC2A1, and VEGFA was determined by immunohistochemistry. A 500-fold cross-validated hazard rate ratio (HRRav) for overall survival was calculated. RESULTS: HIF1α, SLC2A1, and VEGFA expression could be evaluated in 328, 350, and 335 patients, respectively. High SLC2A1 expression was associated with good prognosis (HRRav, 0.67; P (HRR >1)  < 0.01) and high VEGFA expression to poor prognosis (HRRav, 1.84; P (HRR < 1)  = 0.02), also after multivariate analysis including established clinicopathological prognostic variables (HRRav, 0.67; P (HRR > 1)  < 0.01 and HRRav, 1.50; P (HRR < 1)  = 0.02, respectively). SLC2A1 showed prognostic value particularly in patients treated with systemic therapy (P < 0.01), whereas the prognostic value of VEGFA expression was mainly observed in patients not treated with systemic therapy (P < 0.01). Prognosis was especially poor in patients with both low SLC2A1 and high VEGFA expression (P < 0.01). HIF1α expression was not associated with survival. CONCLUSIONS: SLC2A1 and VEGFA expression are prognostic molecular biomarkers for patients with CRCLM with added value to established clinicopathological variables.


Asunto(s)
Transportador de Glucosa de Tipo 1/análisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/cirugía , Factor A de Crecimiento Endotelial Vascular/análisis , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Transportador de Glucosa de Tipo 1/biosíntesis , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Neoplasias Hepáticas/química , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Factor A de Crecimiento Endotelial Vascular/biosíntesis
8.
J Surg Oncol ; 109(3): 255-65, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24249458

RESUMEN

BACKGROUND AND OBJECTIVES: Deregulation of apoptosis related genes may be associated with poor outcome in cancer. Aim of the present study was to investigate the prognostic role of expression levels of apoptosis related proteins in stage II and III colon cancer. METHODS: From tumor samples of 386 stage II and III colon cancer patients, DNA was isolated and tissue microarrays were constructed. Expression of Bcl-2, Bcl-X, BAX, XIAP, Fas, FasL and c-FLIP was evaluated and PCR-based microsatellite instability analysis was performed. RESULTS: High FasL expressing tumors were associated with high disease recurrence rates in stage II colon cancer patients overall, as was low Bcl-X expression in microsatellite stable stage II patients. In stage II patients, a multivariable model based on FasL and Bcl-XL expression revealed a significant association with disease free survival (DFS). In stage III colon cancer patients, low Bcl-2, low BAX and low Fas expression levels were associated with worse outcome. In these patients a multivariable model based on angioinvasion and Bcl-2, Fas and FasL expression was significantly associated with DFS. CONCLUSIONS: Stage II patients with low Bcl-X and high FasL protein expression levels and stage III patients with low Fas, high FasL and low Bcl-2 expression could be considered as high risk for disease recurrence.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/análisis , Biomarcadores de Tumor/análisis , Neoplasias del Colon/química , Neoplasias del Colon/patología , Inestabilidad de Microsatélites , Adulto , Anciano , Anciano de 80 o más Años , Proteínas Reguladoras de la Apoptosis/genética , Biomarcadores de Tumor/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/análisis , Neoplasias del Colon/genética , ADN de Neoplasias/metabolismo , Supervivencia sin Enfermedad , Proteína Ligando Fas/análisis , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Pronóstico , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Recurrencia , Medición de Riesgo , Factores de Riesgo , Análisis de Matrices Tisulares , Proteína Inhibidora de la Apoptosis Ligada a X/análisis , Proteína X Asociada a bcl-2/análisis , Proteína bcl-X/análisis , Receptor fas/análisis
9.
Ann Surg Oncol ; 20 Suppl 3: S348-59, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22711178

RESUMEN

BACKGROUND: Tumor stroma plays an important role in the progression and metastasis of colon cancer. The glycoproteins versican and lumican are overexpressed in colon carcinomas and are associated with the formation of tumor stroma. The aim of the present study was to investigate the potential prognostic value of versican and lumican expression in the epithelial and stromal compartment of Union for International Cancer Control (UICC) stage II and III colon cancer. METHODS: Clinicopathological data and tissue samples were collected from stage II (n = 226) and stage III (n = 160) colon cancer patients. Tissue microarrays were constructed with cores taken from both the center and the periphery of the tumor. These were immunohistochemically stained for lumican and versican. Expression levels were scored on digitized slides. Statistical evaluation was performed. RESULTS: Versican expression by epithelial cells in the periphery of the tumor, i.e., near the invasive front, was correlated to a longer disease-free survival for the whole cohort (P = 0.01), stage III patients only (P = 0.01), stage III patients with microsatellite-instable tumors (P = 0.04), and stage III patients with microsatellite-stable tumors who did not receive adjuvant chemotherapy (P = 0.006). Lumican expression in epithelial cells overall in the tumor was correlated to a longer disease-specific survival in stage II patients (P = 0.05) and to a longer disease-free survival and disease-specific survival in microsatellite-stable stage II patients (P = 0.02 and P = 0.004). CONCLUSIONS: Protein expression of versican and lumican predicted good clinical outcome for stage III and II colon cancer patients, respectively.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Neoplasias del Colon/metabolismo , Sulfato de Queratano/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Versicanos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias del Colon/mortalidad , Neoplasias del Colon/patología , Células Epiteliales/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Lumican , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Clasificación del Tumor , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pronóstico , Células del Estroma/metabolismo , Tasa de Supervivencia , Análisis de Matrices Tisulares
10.
J Pathol ; 226(3): 442-50, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22009326

RESUMEN

Colorectal cancer (CRC) is the second leading cause of cancer death in the western world. The majority of CRCs, which develop from adenoma precursor lesions, show gain of chromosome arm 20q, where BCL2L1 is located. BCL2L1 is an important apoptosis regulating gene that codes for both an anti-apoptotic (Bcl-x(L)) and a pro-apoptotic (Bcl-x(S)) splice variant. The aim of the present study was to investigate whether BCL2L1 contributes to 20q gain-driven colorectal adenoma-to-carcinoma progression. To this end, the functional role of BCL2L1 in cancer-related processes was investigated, and differences in BCL2L1 DNA, mRNA, and protein levels were compared between colorectal adenomas and CRCs, as well as between tumours with and without 20q gain. Down-modulation of BCL2L1 inhibited cell viability and anchorage-independent growth of CRC cells, while invasion was not affected. BCL2L1 DNA copy number and protein expression were increased in CRCs compared to adenomas (p = 0.00005 and p = 0.03, respectively), while mRNA expression was not. Differences in BCL2L1 protein expression were even more pronounced between tumours with and without 20q gain (p = 0.0001). In conclusion, BCL2L1 is functionally involved in several cancer-related processes and its protein expression is associated with 20q gain. This supports a role for 20q gain-dependent expression of BCL2L1 in colorectal adenoma-to-carcinoma progression. However, the absence of a direct correlation between BCL2L1 mRNA and protein expression implies that BCL2L1 protein expression is regulated at the post-transcriptional level by a distinct factor on the 20q amplicon (eg ZNF217, AURKA or miRNAs). Therefore, even though BCL2L1 affects CRC biology in a 20q gain-dependent manner, it is not likely to be a driver of chromosome 20q gain associated adenoma-to-carcinoma progression.


Asunto(s)
Adenoma/genética , Carcinoma/genética , Duplicación Cromosómica/genética , Cromosomas Humanos Par 20/genética , Neoplasias Colorrectales/genética , Proteína bcl-X/fisiología , Transformación Celular Neoplásica/genética , Variaciones en el Número de Copia de ADN/genética , Progresión de la Enfermedad , Humanos , Proteínas de Neoplasias/metabolismo , ARN Mensajero/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
11.
Gut ; 61(11): 1568-75, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22207630

RESUMEN

BACKGROUND AND OBJECTIVE: Progression of a colorectal adenoma to invasive cancer occurs in a minority of adenomas and is the most crucial step in colorectal cancer pathogenesis. In the majority of cases, this is associated with gain of a substantial part of chromosome 20q, indicating that multiple genes on the 20q amplicon may drive carcinogenesis. The aim of this study was to identify genes located on the 20q amplicon that promote progression of colorectal adenoma to carcinoma. DESIGN: Functional assays were performed for 32 candidate driver genes for which a positive correlation between 20q DNA copy number and mRNA expression had been demonstrated. Effects of gene knockdown on cell viability, anchorage-independent growth, and invasion were analysed in colorectal cancer cell lines with 20q gain. Colorectal tumour protein expression was examined by immunohistochemical staining of tissue microarrays. RESULTS: TPX2, AURKA, CSE1L, DIDO1, HM13, TCFL5, SLC17A9, RBM39 and PRPF6 affected cell viability and/or anchorage-independent growth. Chromosome 20q DNA copy number status correlated significantly with TPX2 and AURKA protein levels in a series of colorectal adenomas and carcinomas. Moreover, downmodulation of TPX2 and AURKA was shown to inhibit invasion. CONCLUSION: These data identify TPX2 (20q11) and AURKA (20q13.2) as two genes located on distinct regions of chromosome 20q that promote 20q amplicon-driven progression of colorectal adenoma to carcinoma. Therefore the selection advantage imposed by 20q gain in tumour progression is achieved by gain-of-function of multiple cancer-related genes-knowledge that can be translated into novel tests for early diagnosis of progressive adenomas.


Asunto(s)
Adenocarcinoma/genética , Adenoma/genética , Proteínas de Ciclo Celular/genética , Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , Adenocarcinoma/patología , Adenoma/patología , Aurora Quinasa A , Aurora Quinasas , Biopsia con Aguja , Cromosomas Humanos Par 20/genética , Neoplasias Colorrectales/patología , Variaciones en el Número de Copia de ADN/genética , Progresión de la Enfermedad , Femenino , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , ARN Neoplásico/genética , Muestreo , Sensibilidad y Especificidad , Células Tumorales Cultivadas
12.
Gut ; 61(6): 855-64, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21890811

RESUMEN

BACKGROUND AND OBJECTIVE: Early detection of colon adenomas at high risk of progression and early-stage colorectal cancer (CRC) is an effective approach to reduce CRC death rates. Current screening methods lack specificity as they detect many adenomas that will never progress to CRC. The authors aimed to identify cell surface protein biomarkers with extracellular domains that could be targeted for molecular imaging and discriminate low-risk adenomas and normal colon from high-risk adenomas and CRC. DESIGN: Cell surface proteins of five CRC cell lines were biotinylated, isolated and analysed by in-depth proteomics using gel electrophoresis and nanoliquid chromatography coupled to tandem mass spectrometry. Differential expression in adenomas and CRCs was based on mRNA expression and verified by immunohistochemical staining of tissue microarrays. RESULTS: In total, 2609 proteins were identified in the cell surface fractions. Of these, 44 proteins were selected as promising cell surface candidate biomarkers for adenoma-to-carcinoma progression based on the following criteria: protein identification in at least four out of five cell lines, a predicted (trans)membrane location and increased mRNA expression in CRCs compared to adenomas. Increased protein expression in high-risk adenomas and CRCs compared to low-risk adenomas was confirmed by immunohistochemistry for glucose transporter type 1 (gene symbol SLC2A1; p<0.00001) and prion protein (gene symbol PRNP; p<0.005). CONCLUSION: This study revealed glucose transporter type 1, prion protein and 42 other cell surface candidate biomarkers for adenoma-to-carcinoma progression that could potentially serve as targets for emerging molecular imaging modalities like optical imaging, ¹9F-MRI and positron emission tomography.


Asunto(s)
Adenoma/diagnóstico , Carcinoma/diagnóstico , Neoplasias Colorrectales/diagnóstico , Transportador de Glucosa de Tipo 1/análisis , Priones/análisis , Adenoma/química , Biomarcadores/análisis , Western Blotting , Células CACO-2/química , Carcinoma/química , Línea Celular Tumoral , Neoplasias Colorrectales/química , Variaciones en el Número de Copia de ADN , Progresión de la Enfermedad , Electroforesis en Gel de Poliacrilamida , Células HT29/química , Humanos , Proteínas de Neoplasias/análisis , Proteínas PrPC/análisis , Análisis por Matrices de Proteínas/métodos , Proteómica/métodos
13.
Cancer Res Commun ; 3(11): 2292-2301, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37921412

RESUMEN

Current morphologic features defining advanced adenomas (size ≥10 mm, high-grade dysplasia or ≥25% villous component) cannot optimally distinguish individuals at high risk or low risk of metachronous colorectal cancer (me-CRC), which may result in suboptimal surveillance. Certain DNA copy-number alterations (CNAs) are associated with adenoma-to-carcinoma progression. We aimed to evaluate whether these molecular features can better predict an individual's risk of me-CRC than the morphologic advanced adenoma features.In this nested case-control study, 529 individuals with a single adenoma at first colonoscopy were selected from a Norwegian adenoma cohort. DNA copy-number profiles were determined, by low-coverage whole-genome sequencing. Prevalence of CNAs in advanced and non-advanced adenomas and its association (OR) with me-CRC was assessed. For the latter, cases (with me-CRC) were matched to controls (without me-CRC) on follow-up, age and sex.CNAs associated with adenoma-to-carcinoma progression were observed in 85/267 (32%) of advanced adenomas and in 27/262 (10%) of non-advanced adenomas. me-CRC was statistically significantly associated, also after adjustment for other variables, with age at baseline [OR, 1.14; 95% confidence interval CI), 1.03-1.26; P = 0.012], advanced adenomas (OR, 2.46; 95% CI, 1.50-4.01; P < 0.001) and with the presence of ≥3 DNA copy-number losses (OR, 1.90; 95% CI. 1.02-3.54; P = 0.043).Molecularly-defined high-risk adenomas were associated with me-CRC, but the association of advanced adenoma with me-CRC was stronger. SIGNIFICANCE: Identifying new biomarkers may improve prediction of me-CRC for individuals with adenomas and optimize surveillance intervals to reduce risk of colorectal cancer and reduce oversurveillance of patients with low risk of colorectal cancer. Use of DNA CNAs alone does not improve prediction of me-CRC. Further research to improve risk classification is required.


Asunto(s)
Adenoma , Carcinoma , Neoplasias Colorrectales , Humanos , Neoplasias Colorrectales/diagnóstico , Estudios de Casos y Controles , Adenoma/diagnóstico , ADN
14.
Ann Surg Oncol ; 19 Suppl 3: S682-92, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22311118

RESUMEN

PURPOSE: To investigate the prognostic value of multiple cell cycle-associated proteins in a large series of stage II and III colon cancers. METHODS: From formalin-fixed, paraffin-embedded tumor samples of 386 patients with stage II and III colon cancer, DNA was isolated and tissue microarrays were constructed. Tissue microarray slides were immunohistochemically stained for p21, p27, p53, epidermal growth factor receptor, Her2/Neu, ß-catenin, cyclin D1, Ki-67, thymidylate synthase, and Aurora kinase A (AURKA). Polymerase chain reaction-based microsatellite instability analysis was performed to allow for stratification of protein expression by microsatellite instability status. RESULTS: Overall, low p21, high p53, low cyclin D1, and high AURKA expression were significantly associated with recurrence (P = 0.01, P < 0.01, P = 0.04, and P < 0.01, respectively). In stage II patients who did not receive adjuvant chemotherapy (n = 190), significantly more recurrences were observed in case of low-p21 and high-p53-expressing tumors (P < 0.01 and P = 0.03, respectively). In stage III patients who did not receive chemotherapy, high p53 expression was associated with recurrence (P = 0.02), and in patients who received chemotherapy, high AURKA expression was associated with relapse (P < 0.01). In patients with microsatellite stable tumors, high levels of p53 and AURKA were associated with recurrence (P = 0.01 and P < 0.01, respectively). Multivariate analysis showed p21 (odds ratio 1.6, 95% confidence interval 0.9-2.8) and AURKA (odds ratio 2.7, 95% confidence interval 1.3-5.6) to be independently associated with disease recurrence. CONCLUSIONS: p21, p53, cyclin D1, and AURKA could possibly be used as prognostic markers to identify colon cancer patients with high risk of disease recurrence.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Inestabilidad de Microsatélites , Recurrencia Local de Neoplasia/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Aurora Quinasa A , Aurora Quinasas , Neoplasias del Colon/genética , Neoplasias del Colon/terapia , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Supervivencia sin Enfermedad , Receptores ErbB/metabolismo , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Análisis Multivariante , Recurrencia Local de Neoplasia/genética , Estadificación de Neoplasias , Proteínas Serina-Treonina Quinasas/metabolismo , Curva ROC , Receptor ErbB-2/metabolismo , Timidilato Sintasa/metabolismo , Análisis de Matrices Tisulares , Proteína p53 Supresora de Tumor/metabolismo , beta Catenina/metabolismo
15.
Inflamm Bowel Dis ; 28(9): 1309-1320, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35554535

RESUMEN

BACKGROUND: Patients with primary sclerosing cholangitis (PSC) and inflammatory bowel disease (IBD) run a 10-fold increased risk of developing colorectal cancer (CRC) compared to patients with IBD only. The aim of this study was to perform an extensive screen of known carcinogenic genomic alterations in patients with PSC-IBD, and to investigate whether such changes occur already in nondysplastic mucosa. METHODS: Archival cancer tissue and nondysplastic mucosa from resection specimens of 19 patients with PSC-IBD-CRC were characterized, determining DNA copy-number variations, microsatellite instability (MSI), mutations on 48 cancer genes, and CpG island methylator phenotype (CIMP). Genetic profiles were compared with 2 published cohorts of IBD-associated CRC (IBD-CRC; n = 11) and sporadic CRC (s-CRC; n = 100). RESULTS: Patterns of chromosomal aberrations in PSC-IBD-CRC were similar to those observed in IBD-CRC and s-CRC, MSI occurred only once. Mutation frequencies were comparable between the groups, except for mutations in KRAS, which were less frequent in PSC-IBD-CRC (5%) versus IBD-CRC (38%) and s-CRC (31%; P = .034), and in APC, which were less frequent in PSC-IBD-CRC (5%) and IBD-CRC (0%) versus s-CRC (50%; P < .001). Cases of PSC-IBD-CRC were frequently CIMP positive (44%), at similar levels to cases of s-CRC (34%; P = .574) but less frequent than in cases with IBD-CRC (90%; P = .037). Similar copy number aberrations and mutations were present in matched cancers and adjacent mucosa in 5/15 and 7/11 patients, respectively. CONCLUSIONS: The excess risk of CRC in patients with PSC-IBD was not explained by copy number aberrations, mutations, MSI, nor CIMP status, in cancer tissue, nor in adjacent mucosa. These findings set the stage for further exome-wide and epigenetic studies.


The excessive risk of colorectal carcinoma (CRC) in patients with both primary sclerosing cholangitis and inflammatory bowel disease (IBD) was not explained by an extensive screen of copy number aberrations, mutations, microsatellite instability, and CpG island methylator phenotype status when compared with patients with IBD-CRC and sporadic CRC.


Asunto(s)
Colangitis Esclerosante , Neoplasias Colorrectales , Enfermedades Inflamatorias del Intestino , Colangitis Esclerosante/complicaciones , Colangitis Esclerosante/genética , Colangitis Esclerosante/cirugía , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/genética , Perfil Genético , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/genética , Inestabilidad de Microsatélites
16.
Sci Rep ; 12(1): 12174, 2022 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-35842496

RESUMEN

Secretory leukocyte protease inhibitor (SLPI) is a pleiotropic protein produced by healthy intestinal epithelial cells. SLPI regulates NF-κB activation, inhibits neutrophil proteases and has broad antimicrobial activity. Recently, increased SLPI expression was found in various types of carcinomas and was suggested to increase their metastatic potential. Indeed, we demonstrated that SLPI protein expression in colorectal cancer (CRC) liver metastases and matched primary tumors is associated with worse outcome, suggesting that SLPI promotes metastasis in human CRC. However, whether SLPI plays a role in CRC before distant metastases have formed is unclear. Therefore, we examined whether SLPI expression is associated with prognosis in CRC patients with localized disease. Using a cohort of 226 stage II and 160 stage III CRC patients we demonstrate that high SLPI protein expression is associated with reduced disease recurrence in patients with stage III micro-satellite stable tumors treated with adjuvant chemotherapy, independently of established clinical risk factors (hazard rate ratio 0.54, P-value 0.03). SLPI protein expression was not associated with disease-free survival in stage II CRC patients. Our data suggest that the role of SLPI in CRC may be different depending on the stage of disease. In stage III CRC, SLPI expression may be unfavorable for tumors, whereas SLPI expression may be beneficial for tumors once distant metastases have established.


Asunto(s)
Neoplasias Colorrectales , Inhibidor Secretorio de Peptidasas Leucocitarias , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Regulación de la Expresión Génica , Humanos , Recurrencia Local de Neoplasia , Pronóstico , Inhibidor Secretorio de Peptidasas Leucocitarias/genética , Inhibidor Secretorio de Peptidasas Leucocitarias/metabolismo
17.
Eur J Cancer ; 144: 91-100, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33341450

RESUMEN

AIM: Better stratification of patients with stage II and stage III colon cancer for risk of recurrence is urgently needed. The present study aimed to validate the prognostic value of CDX2 protein expression in colon cancer tissue by routine immunohistochemistry and to evaluate its performance in a head-to-head comparison with tandem mass spectrometry-based proteomics. PATIENT AND METHODS: CDX2 protein expression was evaluated in 386 stage II and III primary colon cancers by immunohistochemical staining of tissue microarrays and by liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis using formalin-fixed paraffin-embedded tissue sections of a matched subset of 23 recurrent and 23 non-recurrent colon cancers. Association between CDX2 expression and disease-specific survival (DSS) was investigated. RESULTS: Low levels of CDX2 protein expression in stage II and III colon cancer as determined by immunohistochemistry was associated with poor DSS (hazard ratio [HR] = 1.97 (95% confidence interval [CI]: 1.26-3.06); p = 0.002). Based on analysis of a selected sample subset, CDX2 prognostic value was more pronounced when detected by LC-MS/MS (HR = 7.56 (95% CI: 2.49-22.95); p < 0.001) compared to detection by immunohistochemistry (HR = 1.60 (95% CI: 0.61-4.22); p = 0.34). CONCLUSION: This study validated CDX2 protein expression as a prognostic biomarker in stage II and III colon cancer, conform previous publications. CDX2 prognostic value appeared to be underestimated when detected by routine immunohistochemistry, probably due to the semiquantitative and subjective nature of this methodology. Quantitative analysis of CDX2 substantially improved its clinical utility as a prognostic biomarker. Therefore, development of routinely applicable quantitative assays for CDX2 expression is needed to facilitate its clinical implementation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/metabolismo , Factor de Transcripción CDX2/metabolismo , Colectomía/mortalidad , Neoplasias del Colon/patología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias del Colon/metabolismo , Neoplasias del Colon/terapia , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia
18.
EBioMedicine ; 70: 103498, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34333237

RESUMEN

BACKGROUND: Recurrence rates after resection of colorectal cancer liver metastases (CRLM) are high and correlate with worse survival. Postoperative circulating tumour DNA (ctDNA) is a promising prognostic biomarker. Focusing on patients with resected CRLM, this study aimed to evaluate the association between the detection of postoperative ctDNA, pathologic response and recurrence-free survival (RFS). METHODS: Twenty-three patients were selected from an ongoing phase-3 trial who underwent resection of RAS-mutant CRLM after induction systemic treatment. CtDNA analysis was performed by droplet digital PCR using blood samples collected at baseline, before and after resection. Pathologic response of CRLM was determined via the Tumour Regression Grading system. FINDINGS: With a median follow-up of 19.6 months, the median RFS for patients with detectable (N = 6, [26%]) and undetectable (N = 17, [74%]) postoperative ctDNA was 4.8 versus 12.1 months, respectively. Among 21 patients with available tumour tissue, pathologic response in patients with detectable compared to undetectable postoperative ctDNA was found in one of six (17%) and 15 of 15 (100%) patients, respectively (p < 0.001). In univariable Cox regression analyses both postoperative detectable ctDNA (HR = 3.3, 95%CI = 1.1-9.6, p = 0.03) and pathologic non-response (HR = 4.6, 95%CI = 1.4-15, p = 0.01) were associated with poorer RFS and were strongly correlated (r = 0.88, p < 0.001). After adjusting for clinical characteristics in pairwise multivariable analyses, postoperative ctDNA status remained associated with RFS. INTERPRETATION: The detection of postoperative ctDNA after secondary resection of CRLM is a promising prognostic factor for RFS and appeared to be highly correlated with pathologic response. FUNDING: None.


Asunto(s)
Biomarcadores de Tumor/sangre , Ácidos Nucleicos Libres de Células/sangre , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/cirugía , Recurrencia Local de Neoplasia/sangre , Complicaciones Posoperatorias/sangre , Anciano , Femenino , Humanos , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/epidemiología , Complicaciones Posoperatorias/epidemiología , Periodo Posoperatorio , Análisis de Supervivencia
19.
Tumour Biol ; 31(2): 89-96, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20358421

RESUMEN

Colorectal adenomas form a biologically and clinically distinct intermediate stage in development of colorectal cancer (CRC) from normal colon epithelium. Only 5% of adenomas progress into adenocarcinomas, indicating that malignant transformation requires other biological alterations than those involved in adenoma formation. The present study aimed to explore which cancer-related biological processes are affected during colorectal adenoma-to-carcinoma progression and to identify key genes within these pathways that can serve as tumor markers for malignant transformation. The activity of 12 cancer-related biological processes was compared between 37 colorectal adenomas and 31 adenocarcinomas, using the pathway analysis tool Gene Set Enrichment Analysis. Expression of six gene sets was significantly increased in CRCs compared to adenomas, representing chromosomal instability, proliferation, differentiation, invasion, stroma activation, and angiogenesis. In addition, 18 key genes were identified for these processes based on their significantly increased expression levels. For AURKA and PDGFRB, increased mRNA expression levels were verified at the protein level by immunohistochemical analysis of a series of adenomas and CRCs. This study revealed cancer-related biological processes whose activities are increased during malignant transformation and identified key genes which may be used as tumor markers to improve molecular characterization of colorectal tumors.


Asunto(s)
Adenoma/genética , Carcinoma/genética , Neoplasias Colorrectales/genética , Adenoma/patología , Aurora Quinasa A , Aurora Quinasas , Carcinoma/patología , Proteínas de Ciclo Celular/análisis , Transformación Celular Neoplásica , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Proteínas Serina-Treonina Quinasas/análisis , Proteínas Proto-Oncogénicas/análisis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/análisis , Quinasa Tipo Polo 1
20.
Oncoimmunology ; 9(1): 1832761, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33101778

RESUMEN

Secretory leukocyte protease inhibitor (SLPI), a pleiotropic protein expressed by healthy intestinal epithelial cells, functions as an inhibitor of NF-κB and neutrophil proteases and exerts antimicrobial activity. We previously showed SLPI suppresses intestinal epithelial chemokine production in response to microbial contact. Increased SLPI expression was recently detected in various types of carcinoma. In addition, accumulating evidence indicates SLPI expression is favorable for tumor cells. In view of these findings and the abundance of SLPI in the colonic epithelium, we hypothesized SLPI promotes colorectal cancer (CRC) growth and metastasis. Here, we aimed to establish whether SLPI expression in CRC is related to clinical outcome. Using a cohort of 507 patients with CRC who underwent resection of liver metastases, we show that high SLPI protein expression in both liver metastases and primary CRC is associated with significantly shorter overall survival after resection of liver metastases. The prognostic value of SLPI in CRC patients with liver metastases implies a role for SLPI in the formation of metastasis of human CRC. Based on the immune regulatory functions of SLPI, we anticipate that expression of SLPI provides tumors with a mechanism to evade infiltration by immune cells.


Asunto(s)
Neoplasias Colorrectales , Neoplasias Hepáticas , Células Epiteliales , Humanos , Pronóstico , Inhibidor Secretorio de Peptidasas Leucocitarias/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA