Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Genet ; 89(1): 128-32, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25827221

RESUMEN

In the Netherlands, the majority of hereditary paragangliomas (PGL) is caused by SDHD, SDHB and SDHAF2 mutations. Founder mutations in SDHD are particularly prevalent, but several SDHB founder mutations have also been described. Here, we describe an extended PGL family with a Dutch founder mutation in SDHB, c.201-4429_287-933del. The proband presented with apparently sporadic head and neck paraganglioma at advanced age. Subsequently, evaluation of the family identified several unaffected mutation carriers, asymptomatic and symptomatic PGL patients, and patients presenting with early-onset malignant pheochromocytoma. The calculated penetrance of the SDHB mutation in this kindred is lower than the risk suggested for SDHB mutations in the literature. This may represent a characteristic of this particular SDHB mutation, but may also be a reflection of the inclusion of relatively large numbers of asymptomatic mutation carriers in this family and adequate statistical correction for ascertainment bias. The low penetrance of SDHB mutations may obscure the hereditary nature of SDHB-linked disease and is important in the counseling of SDHB-linked patients. Risk estimates should preferably be based on the specific mutation involved.


Asunto(s)
Exones , Mutación de Línea Germinal , Paraganglioma/genética , Penetrancia , Feocromocitoma/genética , Eliminación de Secuencia , Succinato Deshidrogenasa/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Femenino , Genotipo , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Paraganglioma/diagnóstico , Paraganglioma/mortalidad , Linaje , Fenotipo , Feocromocitoma/diagnóstico , Feocromocitoma/mortalidad , Adulto Joven
2.
J Med Genet ; 51(5): 283-93, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24556086

RESUMEN

Lynch syndrome (LS) is an autosomal dominant disorder caused by a defect in one of the DNA mismatch repair genes: MLH1, MSH2, MSH6 and PMS2. In the last 15 years, an increasing number of patients have been described with biallelic mismatch repair gene mutations causing a syndrome referred to as 'constitutional mismatch repair-deficiency' (CMMR-D). The spectrum of cancers observed in this syndrome differs from that found in LS, as about half develop brain tumours, around half develop digestive tract cancers and a third develop haematological malignancies. Brain tumours and haematological malignancies are mainly diagnosed in the first decade of life, and colorectal cancer (CRC) and small bowel cancer in the second and third decades of life. Surveillance for CRC in patients with LS is very effective. Therefore, an important question is whether surveillance for the most common CMMR-D-associated cancers will also be effective. Recently, a new European consortium was established with the aim of improving care for patients with CMMR-D. At a workshop of this group held in Paris in June 2013, one of the issues addressed was the development of surveillance guidelines. In 1968, criteria were proposed by WHO that should be met prior to the implementation of screening programmes. These criteria were used to assess surveillance in CMMR-D. The evaluation showed that surveillance for CRC is the only part of the programme that largely complies with the WHO criteria. The values of all other suggested screening protocols are unknown. In particular, it is questionable whether surveillance for haematological malignancies improves the already favourable outcome for patients with these tumours. Based on the available knowledge and the discussions at the workshop, the European consortium proposed a surveillance protocol. Prospective collection of all results of the surveillance is needed to evaluate the effectiveness of the programme.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Trastornos por Deficiencias en la Reparación del ADN/genética , Neoplasias del Sistema Digestivo/diagnóstico , Neoplasias/diagnóstico , Neoplasias Encefálicas/etiología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/tratamiento farmacológico , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Trastornos por Deficiencias en la Reparación del ADN/complicaciones , Humanos , Leucemia/diagnóstico , Mutación , Neoplasias/etiología , Vigilancia de la Población
3.
Br J Cancer ; 105(12): 1912-9, 2011 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-22146830

RESUMEN

BACKGROUND: Birt-Hogg-Dubé (BHD) syndrome is an autosomal dominant condition caused by germline FLCN mutations, and characterised by fibrofolliculomas, pneumothorax and renal cancer. The renal cancer risk, cancer phenotype and pneumothorax risk of BHD have not yet been fully clarified. The main focus of this study was to assess the risk of renal cancer, the histological subtypes of renal tumours and the pneumothorax risk in BHD. METHODS: In this study we present the clinical data of 115 FLCN mutation carriers from 35 BHD families. RESULTS: Among 14 FLCN mutation carriers who developed renal cancer 7 were <50 years at onset and/or had multifocal/bilateral tumours. Five symptomatic patients developed metastatic disease. Two early-stage cases were diagnosed by surveillance. The majority of tumours showed characteristics of both eosinophilic variants of clear cell and chromophobe carcinoma. The estimated penetrance for renal cancer and pneumothorax was 16% (95% minimal confidence interval: 6-26%) and 29% (95% minimal confidence interval: 9-49%) at 70 years of age, respectively. The most frequent diagnosis in families without identified FLCN mutations was familial multiple discoid fibromas. CONCLUSION: We confirmed a high yield of FLCN mutations in clinically defined BHD families, we found a substantially increased lifetime risk of renal cancer of 16% for FLCN mutation carriers. The tumours were metastatic in 5 out of 14 patients and tumour histology was not specific for BHD. We found a pneumothorax risk of 29%. We discuss the implications of our findings for diagnosis and management of BHD.


Asunto(s)
Síndrome de Birt-Hogg-Dubé/genética , Predisposición Genética a la Enfermedad , Neoplasias Renales/genética , Mutación , Neumotórax/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Supresoras de Tumor/genética , Adulto , Anciano , Síndrome de Birt-Hogg-Dubé/complicaciones , Femenino , Humanos , Neoplasias Renales/complicaciones , Masculino , Persona de Mediana Edad , Neumotórax/complicaciones
4.
Clin Genet ; 79(3): 207-18, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21114486

RESUMEN

Previous studies on the counsellees' perception of DNA test results did not clarify whether counsellees were asked about their recollections or interpretations, and focused only on patients' own risks and not on the likelihood that cancer is heritable in the family. We tested differences and correlations of four perception aspects: recollections and interpretations of both cancer risks and heredity likelihood. In a retrospective study, women tested for BRCA1/2 on average, 5 years ago, completed questionnaires about their perception. Participants had received an unclassified variant (n = 76), uninformative (n = 76) or pathogenic mutation (n = 51) result in BRCA1/2. Analyses included t-tests, correlations and structural equation modelling. The counsellees' perception showed to consist of four distinctive phenomena: recollections and interpretations of cancer risks and of heredity likelihood. This distinctiveness was suggested by significant differences between these perception variables. Moderate to strong correlations were found between these variables, suggesting that these differences between variables were consistent. The relationships between these variables were not influenced by actually communicated DNA test results, sociodemographics, medical and pedigree information, or framing of cancer risk questions. The largest differences between recollections and interpretations were found in the unclassified variant group and the smallest in uninformatives. Cancer risks and heredity likelihood correlated least in the pathogenic mutation group. Communication of ambiguous genetic information enlarged the differences. To understand the counsellees' perception of genetic counselling, researchers should study recollections and interpretations of cancer risks and heredity likelihood. Genetic counsellors should explicitly address the counsellees' recollections and interpretations, and be aware of possible inaccuracies.


Asunto(s)
Neoplasias de la Mama , Asesoramiento Genético , Pruebas Genéticas/estadística & datos numéricos , Recuerdo Mental , Neoplasias Ováricas , Percepción , Riesgo , Neoplasias de la Mama/genética , Neoplasias de la Mama/psicología , Femenino , Genes BRCA1 , Genes BRCA2 , Humanos , Persona de Mediana Edad , Modelos Teóricos , Neoplasias Ováricas/genética , Neoplasias Ováricas/psicología , Estudios Retrospectivos , Encuestas y Cuestionarios
5.
Clin Genet ; 80(3): 243-55, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21261604

RESUMEN

Childhood brain tumours may be due to germline bi-allelic mismatch repair (MMR) gene mutations in MLH1, MSH2, MSH6 or PMS2. These mutations can also lead to colorectal neoplasia and haematological malignancies. Here, we review this syndrome and present siblings with early-onset rectal adenoma and papillary glioneural brain tumour, respectively, due to novel germline bi-allelic PMS2 mutations. Identification of MMR protein defects can lead to early diagnosis of this condition. In addition, assays for these defects may help to classify brain tumours for research protocols aimed at targeted therapies.


Asunto(s)
Adenoma/genética , Adenosina Trifosfatasas , Neoplasias Encefálicas/genética , Neoplasias Colorrectales/genética , Enzimas Reparadoras del ADN , Proteínas de Unión al ADN , Mutación de Línea Germinal , Glioma/genética , Adenoma/diagnóstico , Adenoma/patología , Adenosina Trifosfatasas/genética , Edad de Inicio , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patología , Niño , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/patología , Reparación de la Incompatibilidad de ADN , Análisis Mutacional de ADN , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Femenino , Glioma/diagnóstico , Glioma/patología , Heterocigoto , Humanos , Masculino , Repeticiones de Microsatélite , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Linaje , Hermanos , Síndrome , Adulto Joven
6.
Clin Genet ; 79(1): 49-59, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20618355

RESUMEN

Heterozygous fumarate hydratase (FH) germline mutations cause hereditary leiomyomatosis and renal cell cancer (HLRCC), an autosomal dominant syndrome characterized by multiple cutaneous piloleiomyomas, uterine leiomyomas and papillary type 2 renal cancer. The main objective of our study was to evaluate clinical and genetic data from families suspected of HLRCC on a nationwide level. All families referred for FH mutation analysis in the Netherlands were assessed. We performed FH sequence analysis and multiplex ligation-dependent probe amplification. Families with similar FH mutations were examined for haplotype sharing. In 14 out of 33 families, we identified 11 different pathogenic FH germline mutations, including 4 novel mutations and 1 whole-gene deletion. Clinical data were available for 35 FH mutation carriers. Cutaneous leiomyomas were present in all FH mutation carriers older than 40 years of age. Eleven out of 21 female FH mutation carriers underwent surgical treatment for symptomatic uterine leiomyomas at an average of 35 years. Two FH mutation carriers had papillary type 2 renal cancer and Wilms' tumour, respectively. We evaluated the relevance of our findings for clinical practice and have proposed clinical diagnostic criteria, indications for FH mutation analysis and recommendations for management.


Asunto(s)
Carcinoma de Células Renales/genética , Fumarato Hidratasa , Mutación de Línea Germinal , Neoplasias Renales/genética , Leiomiomatosis , Neoplasias Cutáneas/genética , Neoplasias Uterinas/genética , Adolescente , Adulto , Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/enzimología , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Fumarato Hidratasa/genética , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Renales/diagnóstico , Neoplasias Renales/enzimología , Leiomiomatosis/enzimología , Leiomiomatosis/genética , Países Bajos , Linaje , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/enzimología , Síndrome , Neoplasias Uterinas/diagnóstico , Neoplasias Uterinas/enzimología , Adulto Joven
8.
Clin Genet ; 74(2): 145-54, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18510548

RESUMEN

Infantile juvenile polyposis is a rare disease with severe gastrointestinal symptoms and a grave clinical course. Recently, 10q23 microdeletions involving the PTEN and BMPR1A genes were found in four patients with infantile juvenile polyposis. It was hypothesized that a combined and synergistic effect of the deletion of both genes would explain the condition. Subsequently, however, a patient with a larger 10q23 deletion including the same genes but with a mild clinical phenotype was identified. Here, we present four additional patients with 10q23 microdeletions involving the PTEN and BMPR1A genes. The sizes of the deletions were analyzed using single nucleotide polymorphism array analysis. All patients had macrocephaly, dysmorphic features, retardation and congenital abnormalities. One patient developed colorectal cancer. However, only one case had disease onset before 2 years of age and severe symptoms requiring colectomy. No clear correlation was found between ages at onset or severity of gastrointestinal symptoms and the sizes of the deletions. We conclude that patients with 10q23 microdeletions involving the PTEN and BMPR1A genes have variable clinical phenotypes, which cannot be explained merely by the deletion sizes. The phenotypes are not restricted to severe infantile juvenile polyposis but include childhood-onset cases with macrocephaly, retardation, mild gastrointestinal symptoms and possibly early-onset colorectal cancer.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Cromosomas Humanos Par 10 , Enfermedades Gastrointestinales/genética , Poliposis Intestinal/genética , Fosfohidrolasa PTEN/genética , Eliminación de Secuencia , Anomalías Múltiples/genética , Edad de Inicio , Preescolar , Neoplasias Colorrectales/etiología , Femenino , Enfermedades Gastrointestinales/complicaciones , Enfermedades Gastrointestinales/patología , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/genética , Poliposis Intestinal/complicaciones , Poliposis Intestinal/patología , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo
9.
J Clin Invest ; 93(3): 1108-12, 1994 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8132750

RESUMEN

Neonatal severe hyperparathyroidism is a rare life-threatening disorder characterized by very high serum calcium concentrations (> 15 mg/dl). Many cases have occurred in families with familial hypocalciuric hypercalcemia, a benign condition transmitted as a dominant trait. Among several hypothesized relationships between the two syndromes is the suggestion that neonatal severe hyperparathyroidism is the homozygous form of familial hypocalciuric hypercalcemia. To test this hypothesis, we refined the map location of the gene responsible for familial hypocalciuric hypercalcemia on chromosome 3q. Analyses in 11 families defined marker loci closely linked to the gene responsible for familial hypocalciuric hypercalcemia. These loci were then analyzed in four families with parental consanguinity and offspring with neonatal severe hyperparathyroidism. Each individual who was homozygous for loci that are closely linked to the gene responsible for familial hypocalciuric hypercalcemia had neonatal severe hyperparathyroidism. The calculated odds of linkage between these disorders of > 350,000:1 (lod score = 5.56). We conclude that dosage of the gene defect accounts for these widely disparate clinical phenotypes; a single defective allele causes familial hypocalciuric hypercalcemia, while two defective alleles causes neonatal severe hyperparathyroidism.


Asunto(s)
Hipercalcemia/genética , Hiperparatiroidismo/genética , Mutación , Mapeo Cromosómico , Femenino , Haplotipos , Humanos , Masculino , Linaje , Fenotipo
10.
Eur J Cancer ; 43(10): 1556-63, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17532207

RESUMEN

INTRODUCTION: Breast cancer patients with early disease and a natural humoral response to MUC1 have a favourable prognosis, suggesting a possible role of MUC1 antibodies (ab) in controlling haematogenous tumour dissemination and outgrowth. The aim of the study was to evaluate humoral immune responses to MUC1 in women at hereditary high risk of breast cancer to investigate whether this immune response could play a role in the prevention of disease. MATERIALS AND METHODS: CA15.3 (U/mL), and IgG and IgM ab to MUC1 (arbitrary units per mL, Arb-U/mL) were measured in serum samples obtained from 422 women at hereditary high risk of breast/ovarian cancer, of whom 127 BRCA1/2 carriers, attending the Familial Cancer Clinic of the VU University Medical Centre, and from 370 age-matched healthy controls. Serum samples obtained from women who developed breast cancer (N=12) or breast cancer recurrence (N=17), and from women who underwent prophylactic mastectomy (N=12) and had no breast lesions were also tested. RESULTS: CA15.3 ranked significantly higher in mutation carriers than in controls (P=0.03). MUC1 IgG ab levels ranked significantly lower in BRCA1/2 mutation carriers than in controls (P=0.003). MUC1 IgG levels were not significantly different (P=0.53) between women who developed primary breast cancer (median 0.72Arb-U/ml, range 0.52-2.44Arb-U/ml) and women who underwent prophylactic mastectomy and had no breast lesions (median 1.04Arb-U/ml, range 0.43-2.88Arb-U/ml). CONCLUSION: Serum levels of natural IgG ab to MUC1 are lower in BRCA1/2 mutation carriers than in healthy controls. Furthermore, in contrast to previous results in women with sporadic breast cancer, no elevated MUC1 IgG ab were seen in women at hereditary high risk who developed breast cancer. Prophylactic immunotherapy with MUC1 substrates may be a strategy to reduce the risk of breast cancer in BRCA1/2 mutation carriers, strengthening tumour immune surveillance.


Asunto(s)
Anticuerpos/inmunología , Neoplasias de la Mama/inmunología , Genes BRCA1 , Genes BRCA2 , Mucina-1/inmunología , Neoplasias Ováricas/inmunología , Adulto , Anciano , Formación de Anticuerpos/genética , Neoplasias de la Mama/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoglobulina M/inmunología , Persona de Mediana Edad , Mutación/genética , Neoplasias Ováricas/genética
11.
J Med Genet ; 43(8): e41, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16882735

RESUMEN

BACKGROUND: Peutz-Jeghers syndrome (PJS) is caused by germline STK11 mutations and characterised by gastrointestinal polyposis. Although small bowel intussusception is a recognised complication of PJS, risk varies between patients. OBJECTIVE: To analyse the time to onset of intussusception in a large series of PJS probands. METHODS: STK11 mutation status was evaluated in 225 PJS probands and medical histories of the patients reviewed. RESULTS: 135 (60%) of the probands possessed a germline STK11 mutation; 109 (48%) probands had a history of intussusception at a median age of 15.0 years but with wide variability (range 3.7 to 45.4 years). Median time to onset of intussusception was not significantly different between those with identified mutations and those with no mutation detected, at 14.7 years and 16.4 years, respectively (log-rank test of difference, chi(2) = 0.58, with 1df; p = 0.45). Similarly no differences were observed between patient groups on the basis of the type or site of STK11 mutation. CONCLUSIONS: The risk of intussusception in PJS is not influenced by STK11 mutation status.


Asunto(s)
Intususcepción/genética , Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinasas/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo
12.
Ned Tijdschr Geneeskd ; 151(5): 295-8, 2007 Feb 03.
Artículo en Holandés | MEDLINE | ID: mdl-17326472

RESUMEN

Of all forms of cancer, approximately 5% are caused by factors leading to a strong genetic predisposition. DNA diagnosis is currently used in families with hereditary tumour syndromes, such as familial adenomatous polyposis, hereditary non-polyposis colorectal carcinoma (Lynch syndrome), and hereditary breast and ovarian cancer. Those persons who have not inherited the predisposition no longer have to undergo regular examinations. DNA diagnosis for a hereditary predisposition is currently also performed in patients with cancer at a relatively young age, even if the family history is unclear or negative. Consideration of the patient in the context of his or her family is important for both medico-technical and psychosocial reasons. This is true of both diagnostic and presymptomatic DNA diagnosis. For these reasons, the clinical application of the DNA diagnosis of hereditary tumours has become an integral part of the work of the multidisciplinary cancer family clinics of the university medical centres and the cancer centres. Guidelines for the management of hereditary tumours have recently been issued, with criteria for referral to the specialised outpatient clinics.


Asunto(s)
ADN de Neoplasias/análisis , Predisposición Genética a la Enfermedad , Neoplasias/diagnóstico , Neoplasias/genética , Pruebas Genéticas , Humanos , Linaje
13.
J Clin Pathol ; 59(6): 611-7, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16603649

RESUMEN

BACKGROUND: About 5% of all breast cancer cases are attributable to germline mutations in BRCA1 or BRCA2 genes. BRCA mutations in suspected carriers, however, may be missed, which hampers genetic counselling. MATERIALS AND METHODS: Different clinicopathological features were compared between 22 breast cancers from carriers of proved BRCA1 mutations and 604 cancers from sporadic controls. In addition, 5 BRCA2-related breast cancers and 66 breast cancers of untested patients at intermediate risk and 19 breast cancers of untested patients at high risk of hereditary disease on the basis of family history were evaluated. RESULTS: A "probably sporadic" class (age >or=54 years and epidermal growth factor receptor (EGFR) negative; 68% of cases) with a 0% chance of BRCA1-related breast cancer containing 79% of the sporadic cases was yielded by using a decision tree with age, Ki67 and EGFR. A 75% chance of BRCA1-related breast cancer was shown by the "probably BRCA1-related" class (age <54 years and Ki67 >or=25%; 8% of cases) with 82% of the BRCA1-related cases but only 1.4% of the sporadic cases. Most cases at intermediate or high risk of hereditary disease on the basis of family history could be classified with high probability as either probably BRCA1 related or probably sporadic. CONCLUSION: Breast carcinomas can be classified with a high level of certainty as sporadic or related to BRCA1 germline mutations by using a decision tree with age, Ki67 and EGFR. This can be clinically useful in mutation analysis in families with a borderline risk of hereditary disease.


Asunto(s)
Neoplasias de la Mama/genética , Síndromes Neoplásicos Hereditarios/diagnóstico , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Árboles de Decisión , Diagnóstico Diferencial , Receptores ErbB/metabolismo , Femenino , Genes BRCA1 , Genes BRCA2 , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Heterocigoto , Humanos , Técnicas para Inmunoenzimas , Antígeno Ki-67/metabolismo , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Síndromes Neoplásicos Hereditarios/metabolismo , Síndromes Neoplásicos Hereditarios/patología
14.
J Med Genet ; 42(9): 711-9, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16141007

RESUMEN

BACKGROUND: In BRCA2 mutation carriers, increased risks have been reported for several cancer sites besides breast and ovary. As most of the families included in earlier reports were selected on the basis of multiple breast/ovarian cancer cases, it is possible that risk estimates may differ in mutation carriers with a less striking family history. METHODS: In the Netherlands, 139 BRCA2 families with 66 different pathogenic mutations were included in a nationwide study. To avoid testing bias, we chose not to estimate risk in typed carriers, but rather in male and female family members with a 50% prior probability of being a carrier (n = 1811). The relative risk (RR) for each cancer site with the exception of breast and ovarian cancer was determined by comparing observed numbers with those expected, based on Dutch cancer incidence rates. RESULTS: We observed an excess risk for four cancer sites: pancreas (RR 5.9; 95% confidence interval (CI) 3.2 to 10.0), prostate (2.5; 1.6 to 3.8), bone (14.4; 2.9 to 42.1) and pharynx (7.3; 2.0 to 18.6). A small increase was observed for cancer of the digestive tract (1.5; 1.1 to 1.9). Histological verification was available for 46% of the tumours. Nearly all increased risks reached statistical significance for men only. Cancer risks tended to be higher for people before the age of 65 years. Moreover, families with mutations outside the previously defined ovarian cancer cluster region tended to have a higher cancer risk. CONCLUSIONS: We found that BRCA2 carriers are at increased risk for cancers of the prostate and pancreas, and possibly bone and pharynx. Larger databases with extended follow up are needed to provide insight into mutation specific risks of selected carriers in BRCA2 families.


Asunto(s)
Proteína BRCA2/genética , Neoplasias de la Mama/genética , Neoplasias Ováricas/genética , Riesgo , Adulto , Anciano , Neoplasias Óseas/epidemiología , Neoplasias Óseas/genética , Neoplasias de la Mama/epidemiología , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Países Bajos , Neoplasias Ováricas/epidemiología , Neoplasias Pancreáticas/epidemiología , Neoplasias Pancreáticas/genética , Neoplasias Faríngeas/epidemiología , Neoplasias Faríngeas/genética , Neoplasias de la Próstata/epidemiología , Neoplasias de la Próstata/genética
15.
J Med Genet ; 42(9): e54, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16140997

RESUMEN

OBJECTIVE: To investigate the contribution of MYH associated polyposis coli (MAP) among polyposis families in the Netherlands, and the prevalence of colonic and extracolonic manifestations in MAP patients. METHODS: 170 patients with polyposis coli, who previously tested negative for APC mutations, were screened by denaturing gradient gel electrophoresis and direct sequencing to identify MYH germline mutations. RESULTS: Homozygous and compound heterozygous MYH mutations were identified in 40 patients (24%). No difference was found in the percentage of biallelic mutation carriers between patients with 10-99 polyps or 100-1000 polyps (29% in both groups). Colorectal cancer was found in 26 of the 40 patients with MAP (65%) within the age range 21 to 67 years (median 45). Complete endoscopic reports were available for 16 MAP patients and revealed five cases with gastro-duodenal polyps (31%), one of whom also presented with a duodenal carcinoma. Breast cancer occurred in 18% of female MAP patients, significantly more than expected from national statistics (standardised morbidity ratio = 3.75). CONCLUSIONS: Polyp numbers in MAP patients were equally associated with the attenuated and classical polyposis coli phenotypes. Two thirds of the MAP patients had colorectal cancer, 95% of whom were older than 35 years, and one third of a subset of patients had upper gastrointestinal lesions. Endoscopic screening of the whole intestine should be carried out every two years for all MAP patients, starting from age 25-30 years. The frequent occurrence of additional extraintestinal manifestations, such as breast cancer among female MAP patients, should be thoroughly investigated.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , ADN Glicosilasas/genética , Adolescente , Adulto , Anciano , Niño , Neoplasias Colorrectales/genética , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Mutación de Línea Germinal , Humanos , Patrón de Herencia/genética , Masculino , Persona de Mediana Edad , Países Bajos , Fenotipo , Riesgo
16.
Cancer Res ; 56(11): 2621-5, 1996 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-8653707

RESUMEN

The time course of induction of SOS-like stress responses such as enhanced reactivation (ER) and enhanced mutagenesis (EM) has been investigated in UV-C-irradiated skin fibroblasts from a xeroderma pigmentosum (XP) family, using herpes simplex virus type 1 as a probe. Similar ER studies were performed in a Li-Fraumeni syndrome (LFS) family and in a family with a high incidence of breast, ovarian, and colon cancer. In two XP (complementation group B) patients, with a striking absence of skin tumors even at an age of >40 years, only induction of EM was observed, whereas ER was absent (XPER-). The ER- phenotype was inherited from the father, whereas cells from the mother exhibited normal expression of ER and EM. This suggests that the absence of ER is a hereditary trait that is not correlated with a repair-deficient phenotype. Abnormally high levels of ER were observed in UV-C-exposed skin fibroblasts from rive LFS patients. The inheritance of the ER response was studied in one LFS family. High levels of ER were observed only in cells derived from affected individuals carrying one mutated p53 allele, whereas cells from unaffected family members, carrying two wild-type p53 alleles, exhibited normal ER levels. This result shows that abnormally high levels of ER positively correlate with the occurrence of cancer in affected individuals from a LFS family. Interestingly, abnormally high levels of ER were observed in cells from afflicted as well as from unafflicted members of a family with a high incidence of breast, ovarian, colon, and stomach cancer. This suggests that these latter individuals have inherited a mutated, putative predisposing gene, resulting in abnormal expression of ER, but that cancer had not yet developed. The results indicate that the ER response can possibly be used as a prognostic marker to identify carriers in various hereditary cancer-prone syndromes at an early age.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias/genética , Neoplasias Ováricas/genética , Respuesta SOS en Genética , Xerodermia Pigmentosa/genética , Células Cultivadas , ADN Viral/genética , Femenino , Genes Supresores de Tumor , Herpesvirus Humano 1 , Humanos , Masculino , Linaje , Neoplasias Cutáneas/genética , Células Tumorales Cultivadas
17.
Springerplus ; 5(1): 1506, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27652079

RESUMEN

BACKGROUND AND OBJECTIVES: Birt-Hogg-Dubé syndrome is an autosomal dominant disorder characterized by skin fibrofolliculomas, lung cysts, spontaneous pneumothorax and renal cell cancer due to germline folliculin (FLCN) mutations (Menko et al. in Lancet Oncol 10(12):1199-1206, 2009). The aim of this study was to evaluate the incidence of spontaneous pneumothorax in patients with BHD during or shortly after air travel and diving. METHODS: A questionnaire was sent to a cohort of 190 BHD patients and the medical files of these patients were evaluated. The diagnosis of BHD was confirmed by FLCN mutations analysis in all patients. We assessed how many spontaneous pneumothoraces (SP) occurred within 1 month after air travel or diving. RESULTS: In total 158 (83.2 %) patients returned the completed questionnaire. A total of 145 patients had a history of air travel. Sixty-one of them had a history of SP (42.1 %), with a mean of 2.48 episodes (range 1-10). Twenty-four (35.8 %) patients had a history of pneumothorax on both sides. Thirteen patients developed SP < 1 month after air travel (9.0 %) and two patients developed a SP < 1 month after diving (3.7 %). We found in this population of BHD patients a pneumothorax risk of 0.63 % per flight and a risk of 0.33 % per episode of diving. Symptoms possible related to SP were perceived in 30 patients (20.7 %) after air travel, respectively in ten patients (18.5 %) after diving. CONCLUSION: Based on the results presented in this retrospective study, exposure of BHD patients to considerable changes in atmospheric pressure associated with flying and diving may be related to an increased risk for developing a symptomatic pneumothorax. Symptoms reported during or shortly after flying and diving might be related to the early phase of pneumothorax. An individualized advice should be given, taking also into account patients' preferences and needs.

18.
Fam Cancer ; 15(4): 563-70, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26880076

RESUMEN

Familial adenomatous polyposis (FAP) is a dominantly inherited syndrome caused by germline mutations in the APC gene and characterized by the development of multiple colorectal adenomas and a high risk of developing colorectal cancer (CRC). The severity of polyposis is correlated with the site of the APC mutation. However, there is also phenotypic variability within families with the same underlying APC mutation, suggesting that additional factors influence the severity of polyposis. Genome-wide association studies identified several single nucleotide polymorphisms (SNPs) that are associated with CRC. We assessed whether these SNPs are associated with polyp multiplicity in proven APC mutation carriers. Sixteen CRC-associated SNPs were analysed in a cohort of 419 APC germline mutation carriers from 182 families. Clinical data were retrieved from the Dutch Polyposis Registry. Allele frequencies of the SNPs were compared for patients with <100 colorectal adenomas versus patients with ≥100 adenomas, using generalized estimating equations with the APC genotype as a covariate. We found a trend of association of two of the tested SNPs with the ≥100 adenoma phenotype: the C alleles of rs16892766 at 8q23.3 (OR 1.71, 95 % CI 1.05-2.76, p = 0.03, dominant model) and rs3802842 at 11q23.1 (OR 1.51, 95 % CI 1.03-2.22, p = 0.04, dominant model). We identified two risk variants that are associated with a more severe phenotype in APC mutation carriers. These risk variants may partly explain the phenotypic variability in families with the same APC gene defect. Further studies with a larger sample size are recommended to evaluate and confirm the phenotypic effect of these SNPs in FAP.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 8 , Neoplasias Colorrectales/genética , Adenoma/genética , Poliposis Adenomatosa del Colon/genética , Adulto , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Mutación , Polimorfismo de Nucleótido Simple
19.
J Clin Oncol ; 19(20): 4074-80, 2001 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11600610

RESUMEN

PURPOSE: Hereditary nonpolyposis colorectal cancer (HNPCC) is an autosomal dominant disease characterized by the clustering of colorectal cancer, endometrial cancer, and various other cancers. The disease is caused by mutations in DNA-mismatch-repair (MMR) genes, most frequently in MLH1, MSH2, and MSH6. The aims of the present study were to compare the risk of developing colorectal, endometrial, and other cancers between families with the various MMR-gene mutations. PATIENTS AND METHODS: Clinical and pathologic data were collected from 138 families with HNPCC. Mutation analyses were performed for all families. Survival analysis was used to calculate the cumulative risk of developing cancer in the various subsets of relatives. RESULTS: Mutations were identified in 79 families: 34 in MLH1, 40 in MSH2, and five in MSH6. The lifetime risk of developing cancer at any site was significantly higher for MSH2 mutation carriers than for MLH1 mutation carriers (P < .01). The risk of developing colorectal or endometrial cancer was higher in MSH2 mutation carriers than in MLH1 mutation carriers, but the difference was not significant (P = .13 and P = .057, respectively). MSH2 mutation carriers were found to have a significantly higher risk of developing cancer of the urinary tract (P < .05). The risk of developing cancer of the ovaries, stomach, and brain was also higher in the MSH2 mutation carriers than in the MLH1 mutation carriers, but the difference was not statistically significant. CONCLUSION: Pending large prospective studies, the extension of the current surveillance program in MSH2 mutation carriers with the inclusion of the urinary tract should be considered.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Proteínas de Unión al ADN , Mutación , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Adaptadoras Transductoras de Señales , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Disparidad de Par Base/genética , Proteínas Portadoras , Niño , Preescolar , Neoplasias Colorrectales Hereditarias sin Poliposis/epidemiología , Reparación del ADN/genética , Femenino , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Países Bajos/epidemiología , Proteínas Nucleares , Sistema de Registros , Factores de Riesgo , Factores Sexuales
20.
J Clin Pathol ; 58(10): 1091-5, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16189157

RESUMEN

BACKGROUND/AIMS: LKB1 is a tumour suppressor gene that is associated with Peutz-Jeghers syndrome (PJS), a rare autosomal dominant cancer predisposition syndrome. However, germline mutations in the LKB1 gene are found in only about 60% of patients with PJS, suggesting the existence of a second PJS gene. The STRAD gene, encoding an LKB1 interacting protein that activates LKB1, which subsequently leads to polarisation of cells, is an interesting candidate for a second PJS gene and a potential tumour suppressor gene in sporadic carcinomas. METHODS: The involvement of STRAD in 42 PJS associated tumours (sporadic lung, colon, gastric, and ovarian adenocarcinomas) was studied using loss of heterozygosity (LOH) analysis of eight microsatellite markers on chromosome 17, including TP53, BRCA1, and STRAD markers. RESULTS: Loss of the marker near the STRAD locus was seen in 13 of 29 informative cases, including all gastric adenocarcinomas. Specific LOH of the STRAD marker was found in four of 29 informative cases. For these patients all exons and exon-intron boundaries of the STRAD gene were sequenced, but no somatic mutations were identified. Furthermore, no germline STRAD mutations were found in 10 patients with PJS and family members without LKB1 germline mutation. CONCLUSIONS: Despite the frequent occurrence of LOH in the STRAD region, these results indicate that inactivation of the STRAD gene is not essential in the sporadic adenocarcinomas studied, although it is possible that STRAD may be inactivated in different ways. In addition, no evidence was found for the hypothesis that STRAD is a second PJS susceptibility gene.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Adenocarcinoma/genética , Proteínas de Neoplasias/genética , Síndrome de Peutz-Jeghers/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Adenocarcinoma/metabolismo , Cromosomas Humanos Par 17/genética , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , ADN de Neoplasias/genética , Femenino , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Pérdida de Heterocigocidad , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Repeticiones de Microsatélite , Mutación , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Síndrome de Peutz-Jeghers/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA