Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 218
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
CA Cancer J Clin ; 68(3): 217-231, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29485237

RESUMEN

The current understanding of familial colorectal cancer was limited to descriptions of affected pedigrees until the early 1990s. A series of landscape-altering discoveries revealed that there were distinct forms of familial cancer, and most were related to genes previously not known to be involved in human disease. This review largely focuses on advances in our understanding of Lynch syndrome because of the unique relationship of this disease to defective DNA mismatch repair and the clinical implications this has for diagnostics, prevention, and therapy. Recent advances have occurred in our understanding of the epidemiology of this disease, and the advent of broad genetic panels has altered the approach to germline and somatic diagnoses for all of the familial colorectal cancer syndromes. Important advances have been made toward a more complete mechanistic understanding of the pathogenesis of neoplasia in the setting of Lynch syndrome, and these advances have important implications for prevention. Finally, paradigm-shifting approaches to treatment of Lynch-syndrome and related tumors have occurred through the development of immune checkpoint therapies for hypermutated cancers. CA Cancer J Clin 2018;68:217-231. © 2018 American Cancer Society.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/terapia , Adenoma/patología , Transformación Celular Neoplásica , Quimioprevención , Quimioterapia Adyuvante , Colectomía , Colonoscopía , Neoplasias Colorrectales/patología , Neoplasias Colorrectales Hereditarias sin Poliposis/epidemiología , Análisis Mutacional de ADN , Pruebas Genéticas , Mutación de Línea Germinal , Humanos , Análisis de Secuencia de ADN/métodos
2.
Gastroenterology ; 163(5): 1242-1251.e2, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35850198

RESUMEN

BACKGROUND & AIMS: Early-onset colorectal cancer (EOCRC) is a distinct clinical and molecular entity with poor survival outcomes compared with late-onset CRC. Although the incidence of EOCRC is rising, current CRC screening strategies have several limitations in diagnostic performance for EOCRC. In view of this clinical challenge, novel and robust biomarkers for detection of EOCRC are necessary. The aim of this study was to develop a circulating micro RNA (miRNA) signature for the diagnosis of patients with EOCRC. METHODS: A systematic discovery approach by analyzing a large, publicly available, noncoding RNA expression profiling dataset (GSE115513) was used. A panel of miRNAs was identified, which was subsequently validated in blood samples from patients with EOCRC in 2 independent cohorts (n = 149) compared with controls (n = 110) and pre/postoperative plasma specimens (n = 22) using quantitative reverse-transcription polymerase chain reaction assays. RESULTS: In the discovery phase, 4 miRNAs were found to be expressed in blood samples. A combination signature of these 4 miRNAs (miR-193a-5p, miR-210, miR-513a-5p, and miR-628-3p) yielded an area under the curve of 0.92 (95% confidence interval, 0.85-0.96) for identification of EOCRC in the training cohort. The miRNA panel performance was then confirmed in an independent validation cohort (area under the curve, 0.88; 95% confidence interval, 0.82-0.93). Moreover, the miRNA panel robustly identified patients with early-stage EOCRC (P < .001). The decreased expression of miRNAs in postsurgery plasma specimens indicated their tumor specificity. CONCLUSIONS: Our novel miRNA signature for the diagnosis of EOCRC has the potential to identify patients with EOCRC with high accuracy for clinical application in the noninvasive diagnosis of EOCRC.


Asunto(s)
MicroARN Circulante , Neoplasias Colorrectales , MicroARNs , Humanos , Biomarcadores de Tumor/genética , Curva ROC , MicroARNs/genética , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Biopsia Líquida , Perfilación de la Expresión Génica
3.
Gastroenterology ; 162(7): 2063-2085, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35487791

RESUMEN

The gastrointestinal hamartomatous polyposis syndromes are rare, autosomal dominant disorders associated with an increased risk of benign and malignant intestinal and extraintestinal tumors. They include Peutz-Jeghers syndrome, juvenile polyposis syndrome, the PTEN hamartoma tumor syndrome (including Cowden's syndrome and Bannayan-Riley-Ruvalcaba syndrome), and hereditary mixed polyposis syndrome. Diagnoses are based on clinical criteria and, in some cases, confirmed by demonstrating the presence of a germline pathogenic variant. The best understood hamartomatous polyposis syndrome is Peutz-Jeghers syndrome, caused by germline pathogenic variants in the STK11 gene. The management is focused on prevention of bleeding and mechanical obstruction of the small bowel by polyps and surveillance of organs at increased risk for cancer. Juvenile polyposis syndrome is caused by a germline pathogenic variant in either the SMAD4 or BMPR1A genes, with differing clinical courses. Patients with SMAD4 pathogenic variants may have massive gastric polyposis, which can result in gastrointestinal bleeding and/or protein-losing gastropathy. Patients with SMAD4 mutations usually have the simultaneous occurrence of hereditary hemorrhagic telangiectasia (juvenile polyposis syndrome-hereditary hemorrhagic telangiectasia overlap syndrome) that can result in epistaxis, gastrointestinal bleeding from mucocutaneous telangiectasias, and arteriovenous malformations. Germline pathogenic variants in the PTEN gene cause overlapping clinical phenotypes (known as the PTEN hamartoma tumor syndromes), including Cowden's syndrome and related disorders that are associated with an increased risk of gastrointestinal and colonic polyposis, colon cancer, and other extraintestinal manifestations and cancers. Due to the relative rarity of the hamartomatous polyposis syndromes, recommendations for management are based on few studies. This U.S Multi-Society Task Force on Colorectal Cancer consensus statement summarizes the clinical features, assesses the current literature, and provides guidance for diagnosis, assessment, and management of patients with the hamartomatous polyposis syndromes, with a focus on endoscopic management.


Asunto(s)
Neoplasias Colorrectales , Síndrome de Hamartoma Múltiple , Hamartoma , Poliposis Intestinal , Síndromes Neoplásicos Hereditarios , Síndrome de Peutz-Jeghers , Telangiectasia Hemorrágica Hereditaria , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Hemorragia Gastrointestinal , Síndrome de Hamartoma Múltiple/complicaciones , Síndrome de Hamartoma Múltiple/diagnóstico , Síndrome de Hamartoma Múltiple/genética , Humanos , Poliposis Intestinal/complicaciones , Poliposis Intestinal/congénito , Poliposis Intestinal/diagnóstico , Poliposis Intestinal/genética , Pólipos Intestinales , Síndromes Neoplásicos Hereditarios/diagnóstico , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/terapia , Síndrome de Peutz-Jeghers/complicaciones , Síndrome de Peutz-Jeghers/diagnóstico , Síndrome de Peutz-Jeghers/genética
4.
Dig Dis Sci ; 68(9): 3504-3513, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37402979

RESUMEN

In this installment of the "Paradigm Shifts in Perspective" series, the authors, all scientists who have been involved in colorectal cancer (CRC) research for most or all of their careers, have watched the field develop from early pathological descriptions of tumor formation to the current understanding of tumor pathogenesis that informs personalized therapies. We outline how our understanding of the pathogenetic basis of CRC began with seemingly isolated discoveries-initially with the mutations in RAS and the APC gene, the latter of which was initially found in the context of intestinal polyposis, to the more complex process of multistep carcinogenesis, to the chase for tumor suppressor genes, which led to the unexpected discovery of microsatellite instability (MSI). These discoveries enabled the authors to better understand how the DNA mismatch repair (MMR) system not only recognizes DNA damage but also responds to damage by DNA repair or by triggering apoptosis in the injured cell. This work served, in part, to link the earlier findings on the pathogenesis of CRC to the development of immune checkpoint inhibitors, which has been transformative-and curative-for certain types of CRCs and other cancers as well. These discoveries also highlight the circuitous routes that scientific progress takes, which can include thoughtful hypothesis testing and at other times recognizing the importance of seemingly serendipitous observations that substantially change the flow and direction of the discovery process. What has happened over the past 37 years was not predictable when this journey began, but it does speak to the power of careful scientific experimentation, following the facts, perseverance in the face of opposition, and the willingness to think outside of established paradigms.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis , Neoplasias Colorrectales , Humanos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Mutación , Inestabilidad de Microsatélites , Reparación de la Incompatibilidad de ADN/genética
5.
Gastroenterology ; 161(4): 1145-1155.e12, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34119517

RESUMEN

BACKGROUND & AIMS: Incidence and mortality associated with early-age onset colorectal cancer (EAO-CRC) is increasing, prompting professional society recommendations to lower the screening age in average-risk individuals. The yield of screening individuals younger than 50 years is not known. METHODS: A systematic review of 3 databases from inception through July 2020 was performed in all languages that reported colonoscopy findings in average-risk individuals younger than 50 years. The primary outcomes were EAO colorectal neoplasia (CRN) and advanced colorectal neoplasia (aCRN) prevalence. Subgroup analyses were performed based on sex, geographic location, time period, and age, including comparison with those aged 50-59 years. Generalized linear mixed model with random intercept logistic regression and fixed subgroup effects were performed. RESULTS: Of 10,123 unique articles, 17 studies published between 2002 and 2020, including 51,811 average-risk individuals from 4 continents, were included. The pooled rate of EAO-CRN was 13.7% (95% confidence interval [CI], 0.112%-0.168%) and EAO-aCRN was 2.2% (95% CI, 0.016%-0.031%). Prevalence of CRC was 0.05% (95% CI, 0.00029%-0.0008%). Rates of EAO-CRN were higher in men compared with women (relative risk, 1.71%; 95% CI, 1.49%-1.98%), and highest in the United States (15.6%; 95% CI, 12.2%-19.7%) compared with Europe (14.9%; 95% CI, 6.9%-29.3%), East Asia (13.4%; 95% CI, 10.3%-17.2%), and the Middle East (9.8%; 95% CI, 7.8%-12.2%) (P = .04) The rate of EAO-CRN in age groups 45-49 years and 50-59 years was 17.8% (95% CI, 14.5%-21.6%) and 24.8% (95% CI, 19.5%-30.8%), respectively (P = .04). The rate of EAO-aCRN in age group 45-49 years was 3.6% (95% CI, 1.9%-6.7%) and 4.2% (95% CI, 3.2%-5.7%), respectively (P = .69). CONCLUSIONS: The rate of aCRN in individuals aged 45-49 years was similar to the rate observed in individual aged 50-59 years, suggesting that expanding screening to this population could yield a similar impact on colorectal cancer risk reduction.


Asunto(s)
Colonoscopía , Neoplasias Colorrectales/patología , Detección Precoz del Cáncer , Adulto , Edad de Inicio , Neoplasias Colorrectales/epidemiología , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Prevalencia , Medición de Riesgo , Factores de Riesgo
6.
Am J Gastroenterol ; 117(6): 846-864, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35471415

RESUMEN

The gastrointestinal hamartomatous polyposis syndromes are rare, autosomal dominant disorders associated with an increased risk of benign and malignant intestinal and extraintestinal tumors. They include Peutz-Jeghers syndrome, juvenile polyposis syndrome, the PTEN hamartoma tumor syndrome (including Cowden's syndrome and Bannayan-Riley-Ruvalcaba syndrome), and hereditary mixed polyposis syndrome. Diagnoses are based on clinical criteria and, in some cases, confirmed by demonstrating the presence of a germline pathogenic variant. The best understood hamartomatous polyposis syndrome is Peutz-Jeghers syndrome, caused by germline pathogenic variants in the STK11 gene. The management is focused on prevention of bleeding and mechanical obstruction of the small bowel by polyps and surveillance of organs at increased risk for cancer. Juvenile polyposis syndrome is caused by a germline pathogenic variant in either the SMAD4 or BMPR1A genes, with differing clinical courses. Patients with SMAD4 pathogenic variants may have massive gastric polyposis, which can result in gastrointestinal bleeding and/or protein-losing gastropathy. Patients with SMAD4 mutations usually have the simultaneous occurrence of hereditary hemorrhagic telangiectasia (juvenile polyposis syndrome-hereditary hemorrhagic telangiectasia overlap syndrome) that can result in epistaxis, gastrointestinal bleeding from mucocutaneous telangiectasias, and arteriovenous malformations. Germline pathogenic variants in the PTEN gene cause overlapping clinical phenotypes (known as the PTEN hamartoma tumor syndromes), including Cowden's syndrome and related disorders that are associated with an increased risk of gastrointestinal and colonic polyposis, colon cancer, and other extraintestinal manifestations and cancers. Due to the relative rarity of the hamartomatous polyposis syndromes, recommendations for management are based on few studies. This US Multi-Society Task Force on Colorectal Cancer consensus statement summarizes the clinical features, assesses the current literature, and provides guidance for diagnosis, assessment, and management of patients with the hamartomatous polyposis syndromes, with a focus on endoscopic management.


Asunto(s)
Neoplasias Colorrectales , Síndrome de Hamartoma Múltiple , Hamartoma , Poliposis Intestinal , Síndromes Neoplásicos Hereditarios , Síndrome de Peutz-Jeghers , Telangiectasia Hemorrágica Hereditaria , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Hemorragia Gastrointestinal/complicaciones , Hamartoma/complicaciones , Síndrome de Hamartoma Múltiple/complicaciones , Síndrome de Hamartoma Múltiple/diagnóstico , Síndrome de Hamartoma Múltiple/genética , Humanos , Poliposis Intestinal/complicaciones , Poliposis Intestinal/congénito , Poliposis Intestinal/diagnóstico , Poliposis Intestinal/genética , Pólipos Intestinales/complicaciones , Síndromes Neoplásicos Hereditarios/diagnóstico , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/terapia , Síndrome de Peutz-Jeghers/complicaciones , Síndrome de Peutz-Jeghers/diagnóstico , Síndrome de Peutz-Jeghers/genética , Telangiectasia Hemorrágica Hereditaria/complicaciones
7.
Gastrointest Endosc ; 95(6): 1025-1047, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35487765

RESUMEN

The gastrointestinal hamartomatous polyposis syndromes are rare, autosomal dominant disorders associated with an increased risk of benign and malignant intestinal and extraintestinal tumors. They include Peutz-Jeghers syndrome, juvenile polyposis syndrome, the PTEN hamartoma tumor syndrome (including Cowden's syndrome and Bannayan-Riley-Ruvalcaba syndrome), and hereditary mixed polyposis syndrome. Diagnoses are based on clinical criteria and, in some cases, confirmed by demonstrating the presence of a germline pathogenic variant. The best understood hamartomatous polyposis syndrome is Peutz-Jeghers syndrome, caused by germline pathogenic variants in the STK11 gene. The management is focused on prevention of bleeding and mechanical obstruction of the small bowel by polyps and surveillance of organs at increased risk for cancer. Juvenile polyposis syndrome is caused by a germline pathogenic variant in either the SMAD4 or BMPR1A genes, with differing clinical courses. Patients with SMAD4 pathogenic variants may have massive gastric polyposis, which can result in gastrointestinal bleeding and/or protein-losing gastropathy. Patients with SMAD4 mutations usually have the simultaneous occurrence of hereditary hemorrhagic telangiectasia (juvenile polyposis syndrome-hereditary hemorrhagic telangiectasia overlap syndrome) that can result in epistaxis, gastrointestinal bleeding from mucocutaneous telangiectasias, and arteriovenous malformations. Germline pathogenic variants in the PTEN gene cause overlapping clinical phenotypes (known as the PTEN hamartoma tumor syndromes), including Cowden's syndrome and related disorders that are associated with an increased risk of gastrointestinal and colonic polyposis, colon cancer, and other extraintestinal manifestations and cancers. Due to the relative rarity of the hamartomatous polyposis syndromes, recommendations for management are based on few studies. This U.S. Multi-Society Task Force on Colorectal Cancer consensus statement summarizes the clinical features, assesses the current literature, and provides guidance for diagnosis, assessment, and management of patients with the hamartomatous polyposis syndromes, with a focus on endoscopic management.


Asunto(s)
Neoplasias Colorrectales , Síndrome de Hamartoma Múltiple , Hamartoma , Poliposis Intestinal , Síndrome de Peutz-Jeghers , Telangiectasia Hemorrágica Hereditaria , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Hemorragia Gastrointestinal/complicaciones , Síndrome de Hamartoma Múltiple/complicaciones , Síndrome de Hamartoma Múltiple/diagnóstico , Síndrome de Hamartoma Múltiple/genética , Humanos , Poliposis Intestinal/complicaciones , Poliposis Intestinal/congénito , Poliposis Intestinal/diagnóstico , Poliposis Intestinal/genética , Pólipos Intestinales/complicaciones , Síndromes Neoplásicos Hereditarios , Síndrome de Peutz-Jeghers/complicaciones , Síndrome de Peutz-Jeghers/diagnóstico , Síndrome de Peutz-Jeghers/genética , Telangiectasia Hemorrágica Hereditaria/complicaciones
8.
Dig Dis Sci ; 66(3): 855-860, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32300937

RESUMEN

BACKGROUND: Screening for colorectal cancer (CRC) with colonoscopy has been credited for the majority of the decline in mortality over the past 40 years. However, colonoscopy is hampered by frequent interval cancers (ICs) occurring within 36 months after the last examination. Risk factors for IC imply that most misses are amenable to improved colonoscopic techniques. This study had two primary objectives: (1) to determine whether the proportion of persons with ICs has declined in association with improved approaches to the quality of colonoscopy and (2) to determine unrecognized causes for missed diagnoses. METHODS: This is a retrospective, single-center study of persons with CRC having been diagnosed within 6-36 months since the most recent colonoscopy between 2006 and 2015. Participants, ages 50 and 89 years, were evaluated specifically for technical features and findings of the most recent colonoscopy. RESULTS: Thirty-nine persons met the inclusion criteria for IC. Mean age was 69.3 years, range 50-86 years, women were 58%, races included 95% Caucasian, 2.5% Asian, and 2.5% African-American, and history of a first-degree relative with CRC was 26%. Symptoms led to the interval diagnosis in 83%, including iron-deficient anemia (38%), bleeding (25%), abdominal/pelvic pain (18%), and change in bowel habit (2.5%). Preexisting neoplasia was present in 72%. The CRC was located in the proximal colon in 51%. Late-stage disease was present in 45%. When compared to persons with an initial diagnosis of screen-detected late-onset CRC, differences were noted for older age, more women, more with a family history of CRC, and more with late-stage disease. The ratio of interval cancers to new cancers ranged between 6 and 11.5%. Findings indicating a "difficult examination" were observed in 14 cases (36%) compared to "easy or no difficulty" examinations in 25 (65%) cases. In a biennial analysis of IC rates between 2006 and 2015, the percentage of IC remained stable between 6 and 11.5%. CONCLUSIONS: ICs have been stable constituting 6-11.5% of all CRCs. A "difficult examination" was often associated with IC, may be a risk factor for this problem, and may be an important clue warranting further study.


Asunto(s)
Colonoscopía/estadística & datos numéricos , Neoplasias Colorrectales/diagnóstico , Detección Precoz del Cáncer/estadística & datos numéricos , Factores de Tiempo , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
9.
Hum Mutat ; 40(2): 142-161, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30461124

RESUMEN

The epithelial cell adhesion molecule gene (EPCAM, previously known as TACSTD1 or TROP1) encodes a membrane-bound protein that is localized to the basolateral membrane of epithelial cells and is overexpressed in some tumors. Biallelic mutations in EPCAM cause congenital tufting enteropathy (CTE), which is a rare chronic diarrheal disorder presenting in infancy. Monoallelic deletions of the 3' end of EPCAM that silence the downstream gene, MSH2, cause a form of Lynch syndrome, which is a cancer predisposition syndrome associated with loss of DNA mismatch repair. Here, we report 13 novel EPCAM mutations from 17 CTE patients from two separate centers, review EPCAM mutations associated with CTE and Lynch syndrome, and structurally model pathogenic missense mutations. Statistical analyses indicate that the c.499dupC (previously reported as c.498insC) frameshift mutation was associated with more severe treatment regimens and greater mortality in CTE, whereas the c.556-14A>G and c.491+1G>A splice site mutations were not correlated with treatments or outcomes significantly different than random simulation. These findings suggest that genotype-phenotype correlations may be useful in contributing to management decisions of CTE patients. Depending on the type and nature of EPCAM mutation, one of two unrelated diseases may occur, CTE or Lynch syndrome.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Diarrea Infantil/genética , Molécula de Adhesión Celular Epitelial/química , Síndromes de Malabsorción/genética , Modelos Moleculares , Neoplasias Colorrectales Hereditarias sin Poliposis/patología , Diarrea Infantil/patología , Molécula de Adhesión Celular Epitelial/genética , Células Epiteliales/metabolismo , Estudios de Asociación Genética , Humanos , Síndromes de Malabsorción/patología , Proteína 2 Homóloga a MutS/genética , Mutación Missense/genética , Sitios de Empalme de ARN/genética
10.
N Engl J Med ; 374(3): 277-8, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26789877

RESUMEN

Colon cancer has traditionally been treated surgically. However, many cases of colon cancer are systemic at the time of diagnosis, and apparently curative surgery is followed at a later date by tumor recurrence as a consequence of circulating tumor cells before the surgery. Adjuvant medical therapies are designed to prevent recurrences after surgical resection. The current standard for clinical prognostication relies principally on pathological staging. In early-stage colon cancers (stages I and II), all the tumor is contained within the wall of the colon. Less than 10% of patients with stage I disease have a recurrence, and adjuvant chemotherapy is . . .


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/genética , Expresión Génica , Proteínas de Homeodominio/metabolismo , Factor de Transcripción CDX2 , Femenino , Humanos , Masculino
11.
Gastroenterology ; 164(6): 1012-1016, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36997444
12.
Clin Gastroenterol Hepatol ; 17(12): 2600-2602, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30543950

RESUMEN

The proportion of colorectal cancer (CRC) cases in persons younger than age 50, referred to as early onset CRC (EOCRC), has increased from 6% to 11% over the past 25 years, whereas the incidence of CRC has decreased in persons age 50 and older, referred to as late-onset CRC (LOCRC) in the United States.1 It is not known if EOCRC is caused by the same factors that cause LOCRC, or whether there are unique causes that alter the clinical features.2 This study was designed to analyze the clinical and genetic characteristics of EOCRC as presented at a community hospital.


Asunto(s)
Neoplasias Colorrectales/epidemiología , Adolescente , Adulto , Distribución por Edad , Consumo de Bebidas Alcohólicas/epidemiología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/diagnóstico , Reparación de la Incompatibilidad de ADN , Diabetes Mellitus , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto/genética , Proteína 2 Homóloga a MutS/genética , Mutación , Grupos Raciales/estadística & datos numéricos , Sistema de Registros , Distribución por Sexo , Fumar/epidemiología , Adulto Joven
13.
Dig Dis Sci ; 64(11): 3059-3065, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31055721

RESUMEN

BACKGROUND: The incidence of colorectal cancer (CRC) in persons under the age of 50 years (EOCRC) is increasing even as the incidence of CRC in persons over age 50 is decreasing. This has led to recommendations to lower the age of CRC screening to age 45. It is not clear whether EOCRC is identical to CRCs in older patients or whether there are distinctive features between the two groups. AIMS AND METHODS: We reviewed the literature on the clinical and genetic aspects of EOCRC. RESULTS: We found that there is an increased likelihood of a strong genetic basis for EOCRC, but that at least 80% of cases do not come from the known high-penetrance cancer syndromes. Early-onset CRCs tend to occur in the distal colon or rectum, are more likely to be detected due to cancer-related symptoms, appear to be increasing in whites more than non-whites on a population-wide analysis, and are more likely to present in an advanced stage of disease. There are some unique genetic features of EOCRC, including an increased proportion of tumors with LINE-1 hypomethylation, and combined chromosomal and microsatellite stability. CONCLUSIONS: EOCRC deserves additional attention because of the high number of life years at risk with EOCRC, and the implications for earlier CRC screening. Additional focus is needed on determining whether some cases of EOCRC have a unique mechanistic basis.


Asunto(s)
Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Detección Precoz del Cáncer/tendencias , Marcadores Genéticos/genética , Adulto , Factores de Edad , Neoplasias Colorrectales/epidemiología , Detección Precoz del Cáncer/métodos , Pruebas Genéticas/métodos , Pruebas Genéticas/tendencias , Humanos , Incidencia
15.
Gastroenterology ; 153(6): 1634-1646.e8, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28847750

RESUMEN

BACKGROUND & AIMS: Methylation of specific microRNAs (miRNAs) often occurs in an age-dependent manner, as a field defect in some instances, and may be an early event in colitis-associated carcinogenesis. We aimed to determine whether specific mRNA signature patterns (MIR1, MIR9, MIR124, MIR137, MIR34B/C) could be used to identify patients with ulcerative colitis (UC) who are at increased risk for colorectal neoplasia. METHODS: We obtained 387 colorectal tissue specimens collected from 238 patients with UC (152 without neoplasia, 17 with dysplasia, and 69 with UC-associated colorectal cancer [UC-CRC]), from 2 independent cohorts in Japan between 2005 and 2015. We quantified methylation of miRNAs by bisulfite pyrosequencing analysis. We analyzed clinical data to determine whether miRNA methylation patterns were associated with age, location, or segment of the colorectum (cecum, transverse colon, and rectum). Differences in tissue miRNA methylation and expression levels were compared among samples and associated with cancer risk using the Wilcoxon, Mann-Whitney, and Kruskal-Wallis tests as appropriate. We performed a validation study of samples from 90 patients without UC and 61 patients with UC-associated dysplasia or cancer to confirm the association between specific methylation patterns of miRNAs in non-tumor rectal mucosa from patients with UC at risk of UC-CRC. RESULTS: Among patients with UC without neoplasia, rectal tissues had significantly higher levels of methylation levels of MIR1, MIR9, MIR124, and MIR137 than in proximal mucosa; levels of methylation were associated with age and duration of UC in rectal mucosa. Methylation of all miRNAs was significantly higher in samples from patients with dysplasia or CRC compared with samples from patients without neoplasia. Receiver operating characteristic analysis revealed that methylation levels of miRNAs in rectal mucosa accurately differentiated patients with CRC from those without. Methylation of MIR137 in rectal mucosa was an independent risk factor for UC-CRC. Methylation patterns of a set of miRNAs (panel) could discriminate discriminate UC patients with or without dysplasia or CRC in the evaluation cohort (area under the curve, 0.81) and the validation cohort (area under the curve, 0.78). CONCLUSIONS: In evaluation and validation cohorts, we found specific miRNAs to be methylated in rectal mucosal samples from patients with UC with dysplasia or CRC compared with patients without neoplasms. This pattern also associated with patient age and might be used to identify patients with UC at greatest risk for developing UC-CRC. Our findings provide evidence for a field defect in rectal mucosa from patients with UC-CRC.


Asunto(s)
Transformación Celular Neoplásica/genética , Colitis Ulcerosa/genética , Neoplasias Colorrectales/genética , Metilación de ADN , MicroARNs/genética , Transcriptoma , Adulto , Factores de Edad , Área Bajo la Curva , Biopsia , Estudios de Casos y Controles , Colitis Ulcerosa/complicaciones , Colitis Ulcerosa/diagnóstico , Neoplasias Colorrectales/diagnóstico , Femenino , Perfilación de la Expresión Génica/métodos , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Humanos , Masculino , Fenotipo , Valor Predictivo de las Pruebas , Pronóstico , Curva ROC , Medición de Riesgo , Factores de Riesgo
16.
Gastroenterology ; 153(1): 307-323, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28600072

RESUMEN

This document updates the colorectal cancer (CRC) screening recommendations of the U.S. Multi-Society Task Force of Colorectal Cancer (MSTF), which represents the American College of Gastroenterology, the American Gastroenterological Association, and The American Society for Gastrointestinal Endoscopy. CRC screening tests are ranked in 3 tiers based on performance features, costs, and practical considerations. The first-tier tests are colonoscopy every 10 years and annual fecal immunochemical test (FIT). Colonoscopy and FIT are recommended as the cornerstones of screening regardless of how screening is offered. Thus, in a sequential approach based on colonoscopy offered first, FIT should be offered to patients who decline colonoscopy. Colonoscopy and FIT are recommended as tests of choice when multiple options are presented as alternatives. A risk-stratified approach is also appropriate, with FIT screening in populations with an estimated low prevalence of advanced neoplasia and colonoscopy screening in high prevalence populations. The second-tier tests include CT colonography every 5 years, the FIT-fecal DNA test every 3 years, and flexible sigmoidoscopy every 5 to 10 years. These tests are appropriate screening tests, but each has disadvantages relative to the tier 1 tests. Because of limited evidence and current obstacles to use, capsule colonoscopy every 5 years is a third-tier test. We suggest that the Septin9 serum assay (Epigenomics, Seattle, Wash) not be used for screening. Screening should begin at age 50 years in average-risk persons, except in African Americans in whom limited evidence supports screening at 45 years. CRC incidence is rising in persons under age 50, and thorough diagnostic evaluation of young persons with suspected colorectal bleeding is recommended. Discontinuation of screening should be considered when persons up to date with screening, who have prior negative screening (particularly colonoscopy), reach age 75 or have <10 years of life expectancy. Persons without prior screening should be considered for screening up to age 85, depending on age and comorbidities. Persons with a family history of CRC or a documented advanced adenoma in a first-degree relative age <60 years or 2 first-degree relatives with these findings at any age are recommended to undergo screening by colonoscopy every 5 years, beginning 10 years before the age at diagnosis of the youngest affected relative or age 40, whichever is earlier. Persons with a single first-degree relative diagnosed at ≥60 years with CRC or an advanced adenoma can be offered average-risk screening options beginning at age 40 years.


Asunto(s)
Adenoma/diagnóstico , Colonoscopía , Neoplasias Colorrectales/diagnóstico , Detección Precoz del Cáncer/normas , Sangre Oculta , Vigilancia de la Población , Adenoma/genética , Factores de Edad , Colonografía Tomográfica Computarizada , Neoplasias Colorrectales/genética , ADN/análisis , Heces/química , Humanos , Factores de Riesgo , Septinas/sangre , Sigmoidoscopía , Estados Unidos
17.
Gastroenterology ; 152(5): 1217-1237.e3, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27769517

RESUMEN

The use of the fecal occult blood test (FOBT) for colorectal cancer (CRC) screening is supported by randomized trials demonstrating effectiveness in cancer prevention and widely recommended by guidelines for this purpose. The fecal immunochemical test (FIT), as a direct measure of human hemoglobin in stool has a number of advantages relative to conventional FOBT and is increasingly used relative to that test. This review summarizes current evidence for FIT in colorectal neoplasia detection and the comparative effectiveness of FIT relative to other commonly used CRC screening modalities. Based on evidence, guidance statements on FIT application were developed and quality metrics for program implementation proposed.


Asunto(s)
Neoplasias Colorrectales/diagnóstico , Heces/química , Hemoglobinas/análisis , Comités Consultivos , Colonoscopía , Detección Precoz del Cáncer , Humanos , Inmunoquímica , Sangre Oculta , Sensibilidad y Especificidad , Estados Unidos
18.
Gastroenterology ; 152(6): 1605-1614, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28363489

RESUMEN

The US Multi-Society Task Force on Colorectal Cancer, with invited experts, developed a consensus statement and recommendations to assist health care providers with appropriate management of patients with biallelic mismatch repair deficiency (BMMRD) syndrome, also called constitutional mismatch repair deficiency syndrome. This position paper outlines what is known about BMMRD, the unique genetic and clinical aspects of the disease, and reviews the current management approaches to this disorder. This article represents a starting point from which diagnostic and management decisions can undergo rigorous testing for efficacy. There is a lack of strong evidence and a requirement for further research. Nevertheless, providers need direction on how to recognize and care for BMMRD patients today. In addition to identifying areas of research, this article provides guidance for surveillance and management. The major challenge is that BMMRD is rare, limiting the ability to accumulate unbiased data and develop controlled prospective trials. The formation of effective international consortia that collaborate and share data is proposed to accelerate our understanding of this disease.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/terapia , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/terapia , Neoplasias Endometriales/diagnóstico , Neoplasias Hepáticas/diagnóstico , Síndromes Neoplásicos Hereditarios/diagnóstico , Síndromes Neoplásicos Hereditarios/terapia , Vigilancia de la Población , Neoplasias Urológicas/diagnóstico , Alelos , Neoplasias Encefálicas/genética , Neoplasias Colorrectales/genética , Neoplasias Endometriales/genética , Femenino , Asesoramiento Genético , Humanos , Neoplasias Hepáticas/genética , Síndromes Neoplásicos Hereditarios/genética , Neoplasias Urológicas/genética
19.
J Transl Med ; 16(1): 5, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29329588

RESUMEN

BACKGROUND: To screen tumors with microsatellite instability (MSI) arising due to DNA mismatch repair deficiency (dMMR), a panel of five quasi-monomorphic mononucleotide-repeat markers amplified in a multiplex PCR (Pentaplex) are commonly used. In spite of its several strengths, the pentaplex assay is not robust at detecting the loss of MSH6-deficiency (dMSH6). In order to overcome this challenge, we designed this study to develop and optimize a panel of four quasi-monomorphic mononucleotide-repeat markers (Tetraplex) for identifying solid tumors with dMMR, especially dMSH6. METHODS: To improve the sensitivity for tumors with dMMR, we established a quasi-monomorphic variant range (QMVR) of 3-4 bp for the four Tetraplex markers. Thereafter, to confirm the accuracy of this assay, we examined 317 colorectal cancer (CRC) specimens, comprising of 105 dMMR [45 MutL homolog (MLH)1-deficient, 45 MutS protein homolog (MSH)2-deficient, and 15 MSH6-deficient tumors] and 212 MMR-proficient (pMMR) tumors as a test set. In addition, we analyzed a cohort of 138 endometrial cancers (EC) by immunohistochemistry to determine MMR protein expression and validation of our new MSI assay. RESULTS: Using the criteria of ≥ 1 unstable markers as MSI-positive tumor, our assay resulted in a sensitivity of 97.1% [95% confidence interval (CI) = 91.9-99.0%] for dMMR, and a specificity of 95.3% (95% CI = 91.5-97.4%) for pMMR CRC specimens. Among the 138 EC specimens, 41 were dMMR according to immunohistochemistry. Herein, our Tetraplex assay detected dMMR tumors with a sensitivity of 92.7% (95% CI = 80.6-97.5%) and a specificity of 97.9% (95% CI = 92.8-99.4%) for pMMR tumors. With respect to tumors with dMSH6, in the CRC-validation set, Tetraplex detected dMSH6 tumors with a sensitivity of 86.7% (13 of 15 dMSH6 CRCs), which was subsequently validated in the EC test set as well (sensitivity, 75.0%; 6 of 8 dMSH6 ECs). CONCLUSIONS: Our newly optimized Tetraplex system will help offer a robust and highly sensitive assay for the identification of dMMR in solid tumors.


Asunto(s)
Biomarcadores de Tumor/genética , Reparación de la Incompatibilidad de ADN/genética , Repeticiones de Microsatélite/genética , Neoplasias/genética , Alelos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , ADN/genética , Células Germinativas/metabolismo , Humanos , Reproducibilidad de los Resultados
20.
Gut ; 66(4): 654-665, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-26701878

RESUMEN

BACKGROUND AND AIMS: Distant metastasis is a major cause of deaths in patients with colorectal cancer (CRC), which is partly due to lack of robust metastasis-predictive biomarkers. In spite of the important function of microRNA (miR)-203 in cancer metastasis, its clinical significance in CRC metastasis remains unknown. Here, we evaluated the potential role of serum miR-203 as a non-invasive biomarker for CRC metastasis. METHODS: MiR-203 expression was quantified by quantitative reverse-transcription PCR in 58 pairs of primary CRC (pCRC) and corresponding matched liver metastasis (LM), as well as 186 serum and 154 matched tissue specimens from patients with CRC in cohort 1. Next, we performed validation of miR-203 levels in serum from 144 patients with CRC in an independent cohort (cohort 2). Mouse models of CRC-associated metastases were established to identify the source of circulating miR-203. Expression patterns of miR-203 in tissues were determined by in situ hybridisation. RESULTS: MiR-203 expression was significantly upregulated in LM compared with matched pCRC tissues. Serum miR-203 levels were significantly upregulated in a stage-dependent manner, and high miR-203 expression was associated with poor survival in patients with CRC in both patient cohorts. Increased miR-203 levels in serum indicated high risk for poor prognosis (HR=2.1), as well as metastasis to lymph nodes (OR=2.5), liver (OR=6.2), peritoneum (OR=7.2) and distant organs (OR=4.4). Serum miR-203 levels were significantly higher in animals with liver or systemic metastasis compared with controls. CONCLUSIONS: High levels of serum miR-203 associate with poor survival and metastasis, suggesting it to be a promising non-invasive prognostic and metastasis-predictive biomarker in patients with CRC.


Asunto(s)
Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/sangre , Neoplasias Hepáticas/secundario , MicroARNs/sangre , Neoplasias Peritoneales/sangre , Neoplasias Peritoneales/secundario , Anciano , Animales , Biomarcadores de Tumor/sangre , Neoplasias Colorrectales/genética , Femenino , Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Metástasis Linfática , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Estadificación de Neoplasias , Neoplasias Peritoneales/genética , Valor Predictivo de las Pruebas , Pronóstico , Tasa de Supervivencia , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA